JPWO2016027781A1 - Salts and crystals of monocyclic pyridine derivatives - Google Patents

Salts and crystals of monocyclic pyridine derivatives Download PDF

Info

Publication number
JPWO2016027781A1
JPWO2016027781A1 JP2015560425A JP2015560425A JPWO2016027781A1 JP WO2016027781 A1 JPWO2016027781 A1 JP WO2016027781A1 JP 2015560425 A JP2015560425 A JP 2015560425A JP 2015560425 A JP2015560425 A JP 2015560425A JP WO2016027781 A1 JPWO2016027781 A1 JP WO2016027781A1
Authority
JP
Japan
Prior art keywords
ppm
methoxyethoxy
methyl
oxy
indole
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
JP2015560425A
Other languages
Japanese (ja)
Other versions
JP5925978B1 (en
Inventor
俊亮 尾崎
俊亮 尾崎
賢史 吉田
賢史 吉田
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eisai R&D Management Co Ltd
Original Assignee
Eisai R&D Management Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eisai R&D Management Co Ltd filed Critical Eisai R&D Management Co Ltd
Application granted granted Critical
Publication of JP5925978B1 publication Critical patent/JP5925978B1/en
Publication of JPWO2016027781A1 publication Critical patent/JPWO2016027781A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C55/00Saturated compounds having more than one carboxyl group bound to acyclic carbon atoms
    • C07C55/02Dicarboxylic acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C55/00Saturated compounds having more than one carboxyl group bound to acyclic carbon atoms
    • C07C55/02Dicarboxylic acids
    • C07C55/10Succinic acid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Abstract

医薬品の原薬として利用可能性を有する、コハク酸およびマレイン酸からなる群より選択される有機カルボン酸と、式(I)で表される化合物とからなる塩またはその結晶を提供する。【化1】Provided is a salt or crystal thereof comprising an organic carboxylic acid selected from the group consisting of succinic acid and maleic acid, and a compound represented by formula (I), which can be used as an active pharmaceutical ingredient. [Chemical 1]

Description

本発明は、FGFR阻害作用を有する単環ピリジン誘導体の塩およびその結晶に関する。   The present invention relates to a salt of a monocyclic pyridine derivative having an FGFR inhibitory action and a crystal thereof.

FGF(線維芽細胞成長因子:fibroblast growth factor)は、細胞増殖、細胞遊走、細胞浸潤、細胞生存、分化誘導、創傷治癒、血管新生、など多彩な生理機能を制御する成長因子として知られている。
FGFは受容体チロシンキナーゼであるFGF受容体(FGFR:FGFR1、FGFR2、FGFR3、FGFR4)を介して、さまざまな生理機能を制御している。FGFRは細胞外ドメイン、膜貫通ドメインおよび細胞内チロシンキナーゼドメインの3種類から構成されている。FGFがFGFR細胞外ドメインと結合することで、受容体の二量体が形成される。その後、細胞内チロシンキナーゼが活性化された後、主にMAPK(mitogen-activated protein kinase)/ERK(extracellular signal-regulated kinase)経路やPI3K(phosphatidylinositol 3-kinase)/Akt経路を介して細胞内シグナルが伝達される。
FGF (fibroblast growth factor) is known as a growth factor that controls various physiological functions such as cell proliferation, cell migration, cell invasion, cell survival, differentiation induction, wound healing, and angiogenesis. .
FGF regulates various physiological functions through FGF receptors (FGFR: FGFR1, FGFR2, FGFR3, FGFR4) which are receptor tyrosine kinases. FGFR is composed of three types: an extracellular domain, a transmembrane domain, and an intracellular tyrosine kinase domain. FGF binds to the FGFR extracellular domain to form a receptor dimer. Then, after intracellular tyrosine kinases are activated, intracellular signals mainly via MAPK (mitogen-activated protein kinase) / ERK (extracellular signal-regulated kinase) pathway or PI3K (phosphatidylinositol 3-kinase) / Akt pathway. Is transmitted.

一方、FGF産生亢進、FGFR遺伝子増幅、FGFR過剰発現、FGFR融合蛋白形成、FGFR変異などに伴ってFGF/FGFRシグナル異常が誘発される結果、乳癌、膀胱癌、EMS(8p11 myeloproliferative syndrome)、胃癌、子宮内膜癌、前立腺癌等、さまざまな癌が引き起こされることが報告されている(非特許文献1)。さらに、FGF/FGFRシグナル異常を伴うがんとして、非小細胞肺癌、小細胞肺癌、卵巣癌、肉腫、大腸癌、メラノーマ、膠芽細胞腫、星状細胞腫または頭頸部癌(非特許文献2、3)、甲状腺癌(非特許文献4)、膵臓癌(非特許文献5、6)、肝臓癌(非特許文献7)、皮膚癌(非特許文献8)、腎癌(非特許文献9)、肺扁平上皮癌(非特許文献10、11、12)などが報告されている。   On the other hand, as a result of FGF / FGFR signal abnormality induced by FGF production enhancement, FGFR gene amplification, FGFR overexpression, FGFR fusion protein formation, FGFR mutation, etc., breast cancer, bladder cancer, EMS (8p11 myoproliferative syndrom), gastric cancer, It has been reported that various cancers such as endometrial cancer and prostate cancer are caused (Non-patent Document 1). Furthermore, non-small cell lung cancer, small cell lung cancer, ovarian cancer, sarcoma, colon cancer, melanoma, glioblastoma, astrocytoma, or head and neck cancer as a cancer with FGF / FGFR signal abnormality (Non-patent Document 2) 3), thyroid cancer (Non-patent document 4), pancreatic cancer (Non-patent documents 5 and 6), liver cancer (Non-patent document 7), skin cancer (Non-patent document 8), renal cancer (Non-patent document 9) And squamous cell carcinoma of the lung (Non-Patent Documents 10, 11, and 12) have been reported.

また、内皮細胞において、FGF/FGFRシグナルは、VEGF(vascular endothelial growth factor)/KDR(kinase-insert domain-containing receptor)シグナルと並んで主要な血管新生シグナルでもあり、また癌間質細胞(繊維芽細胞)と癌細胞との相互作用にも関与が報告されている(非特許文献1)。
したがって、FGF/FGFRシグナルを標的とするFGFR阻害剤は、FGF/FGFRシグナル異常を伴う癌において、そのシグナル異常に対する抑制作用や血管新生シグナルの抑制作用などに基づく抗腫瘍剤として期待されている。最近になって、他のシグナルの交絡的影響(Confronting effect)を受け難いと考えられる選択的FGFR阻害剤、例えば、本発明に係る化合物とは構造上明らかに異なっている、FGFR1、FGFR2およびFGFR3の選択的FGFR阻害剤が報告されている。しかし、ヒトの抗腫瘍剤としての開発において、選択的FGFR阻害剤は、FGF/FGFRシグナルとVEGF/KDRシグナルの両者を同時に標的とする抗腫瘍剤に遅れを取っており、未だ、市販されていない(非特許文献13、14、特許文献1、2)。特許文献3には、ピリミジン誘導体が開示されているが、FGF/FGFRシグナルのシグナル異常に対する抑制作用は開示されていない。特許文献4には、VEGFおよびFGFにより誘導される血管新生を抑制するピリジン誘導体またはピリミジン誘導体が開示されている。しかしながら、これらの文献には本発明に係る化合物は開示されていない。
In endothelial cells, FGF / FGFR signal is a major angiogenesis signal along with VEGF (vascular endothelial growth factor) / KDR (kinase-insert domain-containing receptor) signal, and also cancer stromal cells (fibroblasts). (Cell non-patent document 1).
Therefore, an FGFR inhibitor targeting FGF / FGFR signal is expected as an antitumor agent based on a suppressive action on the abnormal signal or angiogenic signal in cancer with abnormal FGF / FGFR signal. Recently, selective FGFR inhibitors that are considered to be less susceptible to other signal confronting effects, such as FGFR1, FGFR2 and FGFR3, which are clearly different in structure from the compounds of the present invention. Selective FGFR inhibitors have been reported. However, in the development of human anti-tumor agents, selective FGFR inhibitors lag behind anti-tumor agents that simultaneously target both FGF / FGFR signals and VEGF / KDR signals, and are still commercially available. None (Non-Patent Documents 13 and 14, Patent Documents 1 and 2). Patent Document 3 discloses a pyrimidine derivative, but does not disclose an inhibitory action on signal abnormality of FGF / FGFR signal. Patent Document 4 discloses a pyridine derivative or a pyrimidine derivative that suppresses angiogenesis induced by VEGF and FGF. However, these documents do not disclose the compounds according to the present invention.

国際公開第2008/075068号International Publication No. 2008/075068 国際公開第2006/000420号International Publication No. 2006/000420 国際公開第2002/032872号International Publication No. 2002/032872 国際公開第2004/020434号International Publication No. 2004/020434

Nicholas et al.,”Fibroblast growth factor signalling:from development to cancer”, Nature Reviews Cancer. 2010;10:116-129Nicholas et al., “Fibroblast growth factor signaling: from development to cancer”, Nature Reviews Cancer. 2010; 10: 116-129 Joergen WESCHE et al., Fibroblast growth factors and their receptors in cancer, Biochem J. 2011:437;199-213Joergen WESCHE et al., Fibroblast growth factors and their receptors in cancer, Biochem J. 2011: 437; 199-213 Gennaro Daniele et al., FGF Receptor Inhibitors: Role in Cancer Therapy, Curr Oncol Rep. 2012;14:111-119Gennaro Daniele et al., FGF Receptor Inhibitors: Role in Cancer Therapy, Curr Oncol Rep. 2012; 14: 111-119 Rosanne St. Bernard et al., Fibroblast Growth Factor Receptors as Molecular Targets in Thyroid Carcinoma, Endocrinology. 2005;146:1145-1153Rosanne St. Bernard et al., Fibroblast Growth Factor Receptors as Molecular Targets in Thyroid Carcinoma, Endocrinology. 2005; 146: 1145-1153 Toshiyuki Ishiwata et al., Enhanced Expression of Fibroblast Growth Factor Receptor 2 IIIc Promotes Human Pancreatic Cancer Cell Proliferation, Am J Pathol. 2012;180:1928-1941Toshiyuki Ishiwata et al., Enhanced Expression of Fibroblast Growth Factor Receptor 2 IIIc Promotes Human Pancreatic Cancer Cell Proliferation, Am J Pathol. 2012; 180: 1928-1941 G Chen et al., Inhibition of endogenous SPARC enhances pancreatic cancer cell growth: modulation by FGFR1-III isoform expression, Br J Cancer. 2010;102:188-195G Chen et al., Inhibition of inherent SPARC enhances pancreatic cancer cell growth: modulation by FGFR1-III isoform expression, Br J Cancer. 2010; 102: 188-195 Dorothy M. French et al., Targeting FGFR4 Inhibits Hepatocellular Carcinoma in Preclinical Mouse Models, PLoS One. 2012;7:e36713Dorothy M. French et al., Targeting FGFR4 Inhibits Hepatocellular Carcinoma in Preclinical Mouse Models, PLoS One. 2012; 7: e36713 Armelle Logie et al., Activating mutations of the tyrosine kinase receptor FGFR3 are associated with benign skin tumors in mice and humans, Hum Mol Genet 2005;14:1153-1160Armelle Logie et al., Activating mutations of the tyrosine kinase receptor FGFR3 are associated with benign skin tumors in mice and humans, Hum Mol Genet 2005; 14: 1153-1160 Tsimafeyeu I et al., Overexpression of fibroblast growth factor receptors FGFR1 and FGFR2 in renal cell carcinoma, Scand J Urol Nephrol 2011;45:190-195Tsimafeyeu I et al., Overexpression of fibroblast growth factor receptors FGFR1 and FGFR2 in renal cell carcinoma, Scand J Urol Nephrol 2011; 45: 190-195 Jonathan Weiss et al., Frequent and Focal FGFR1 Amplification Associates with Therapeutically Tractable FGFR1 Dependency in Squamous Cell Lung Cancer, Sci Transl Med. 2010;2:issue62 62-93Jonathan Weiss et al., Frequent and Focal FGFR1 Amplification Associates with Therapeutically Tractable FGFR1 Dependency in Squamous Cell Lung Cancer, Sci Transl Med. 2010; 2: issue62 62-93 Hidefumi Sasaki et al., Increased FGFR1 copy number in lung squamous cell carcinomas, Mol Med Report. 2012;5:725-728Hidefumi Sasaki et al., Increased FGFR1 copy number in lung squamous cell carcinomas, Mol Med Report. 2012; 5: 725-728 The Cancer Genome Atlas Research Network, Comprehensive genomic characterization of squamous cell lung cancers, Nature 2012;489:519-525The Cancer Genome Atlas Research Network, Comprehensive genomic characterization of squamous cell lung cancers, Nature 2012; 489: 519-525 Paul R Gavine et al., AZD4547:An Orally Bioavailable, Potent, and Selective Inhibitor of the Fibroblast Growth Factor Receptor Tyrosine Kinase Family, Cancer Res. 2012;72:2045-2056Paul R Gavine et al., AZD4547: An Orally Bioavailable, Potent, and Selective Inhibitor of the Fibroblast Growth Factor Receptor Tyrosine Kinase Family, Cancer Res. 2012; 72: 2045-2056 Vito Guagnano et al., Discovery of 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea (NVP-BGJ398), A Potent and Selective Inhibitor of the Fibroblast Growth Factor Receptor Family of Receptor Tyrosine Kinase, J Med Chem. 2011;54:7066-7083Vito Guagnano et al., Discovery of 3- (2,6-Dichloro-3,5-dimethoxy-phenyl) -1- {6- [4- (4-ethyl-piperazin-1-yl) -phenylamino] -pyrimidin -4-yl} -1-methyl-urea (NVP-BGJ398), A Potent and Selective Inhibitor of the Fibroblast Growth Factor Receptor Family of Receptor Tyrosine Kinase, J Med Chem. 2011; 54: 7066-7083

下記式(I)で表される化合物(5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド、以下化合物(I)という。)は、FGFR1、FGFR2およびFGFR3阻害作用を有している。
一般に、医薬品として用いられる化合物およびその塩ならびにそれらの結晶の物性は、薬物のバイオアベイラビリティー、原薬の純度、製剤の処方などに大きな影響を与える。したがって、本発明の課題は、医薬品の原薬としての利用可能性を有する化合物(I)の塩およびその結晶を提供することにある。
A compound represented by the following formula (I) (5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy ) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide, hereinafter referred to as Compound (I)) has FGFR1, FGFR2 and FGFR3 inhibitory activity.
In general, the properties of compounds used as pharmaceuticals and salts thereof and crystals thereof have a great influence on the bioavailability of drugs, the purity of drug substances, the formulation of pharmaceuticals, and the like. Accordingly, an object of the present invention is to provide a salt of Compound (I) and a crystal thereof having applicability as an active pharmaceutical ingredient.

本発明者らは、化合物(I)について、上記事情に鑑み鋭意研究を重ねた結果、化合物(I)の塩およびその結晶を見出し、本発明を完成した。   As a result of intensive studies on the compound (I) in view of the above circumstances, the present inventors have found a salt of the compound (I) and crystals thereof and completed the present invention.

すなわち、本発明は、以下の[1]〜[17]を提供する。
[1]下記式(I)で表される5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドと、コハク酸またはマレイン酸からなる塩、
[2]コハク酸からなる塩である、[1]記載の塩、
[3]マレイン酸からなる塩である、[1]記載の塩、
[4]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩である、[2]記載の塩、
[5]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 0.5コハク酸塩である、[2]記載の塩、
[6]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド マレイン酸塩である、[1]記載の塩、
[7]下記式(I)で表される5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドと、コハク酸またはマレイン酸からなる塩の結晶、
[8]下記式(I)で表される5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドと、コハク酸からなる塩の結晶、
[9]下記式(I)で表される5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドと、マレイン酸からなる塩の結晶、
[10]粉末X線回折において、回折角度(2θ±0.2°)22.4°に回折ピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩の結晶、
[11]粉末X線回折において、回折角度(2θ±0.2°)22.4°、25.3°および23.3°に回折ピークを有する、[10]の結晶、
[12]粉末X線回折において、回折角度(2θ±0.2°)19.8°に回折ピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 0.5コハク酸塩の結晶(α)、
[13]粉末X線回折において、回折角度(2θ±0.2°)20.1°に回折ピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド マレイン酸塩の結晶、
[14]13C固体NMRスペクトルにおいて、化学シフト(±0.5ppm)108.5ppm、155.1ppmおよび179.9ppmにピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩の結晶、
[15]13C固体NMRスペクトルにおいて、化学シフト(±0.5ppm)27.1ppm、34.8ppm、108.5ppm、155.1ppmおよび179.9ppmにピークを有する、[14]の結晶、
[16]図1の粉末X線回折パターンを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩の結晶、
[17] [1]〜[16]のいずれか一つに記載の塩または結晶を有効成分として含有する医薬組成物。
That is, the present invention provides the following [1] to [17].
[1] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} represented by the following formula (I) A salt comprising oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and succinic acid or maleic acid,
[2] The salt according to [1], which is a salt composed of succinic acid,
[3] The salt according to [1], which is a salt consisting of maleic acid,
[4] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxy Ethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 succinate, salt according to [2],
[5] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxy Ethoxy) -N-methyl-1H-indole-1-carboxamide 0.5 succinate, salt according to [2],
[6] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxy The salt according to [1], which is ethoxy) -N-methyl-1H-indole-1-carboxamide maleate,
[7] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} represented by the following formula (I) Crystals of a salt consisting of (oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and succinic acid or maleic acid,
[8] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} represented by the following formula (I) Crystals of a salt consisting of oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and succinic acid,
[9] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} represented by the following formula (I) Oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and a crystal of a salt consisting of maleic acid,
[10] 5-({2-[({4- [1- (2-hydroxyethyl) piperidine] having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 22.4 ° in powder X-ray diffraction. -4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 succinate crystals,
[11] In the powder X-ray diffraction, the crystals of [10] having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 22.4 °, 25.3 °, and 23.3 °,
[12] 5-({2-[({4- [1- (2-hydroxyethyl) piperidine] having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 19.8 ° in powder X-ray diffraction -4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 0.5 succinate crystals (α ),
[13] 5-({2-[({4- [1- (2-hydroxyethyl) piperidine] having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 20.1 ° in powder X-ray diffraction -4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide maleate crystals,
[14] In a 13 C solid state NMR spectrum, 5-({2-[({4- [1- (2) having peaks at chemical shifts (± 0.5 ppm) of 108.5 ppm, 155.1 ppm and 179.9 ppm. -Hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 Succinic acid Salt crystals,
[15] Crystal of [14] having peaks at chemical shift (± 0.5 ppm) 27.1 ppm, 34.8 ppm, 108.5 ppm, 155.1 ppm and 179.9 ppm in 13 C solid state NMR spectrum,
[16] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridine-4- having the powder X-ray diffraction pattern of FIG. Yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 succinate crystals,
[17] A pharmaceutical composition comprising the salt or crystal according to any one of [1] to [16] as an active ingredient.

本発明により提供される、化合物(I)の塩およびその結晶は、実施例で示されるような性状であり、また、後述する試験例のデータに示されるような吸湿性の特徴を有し、医薬品の原薬としての利用可能性を有する。   The salt of compound (I) and the crystal thereof provided by the present invention have properties as shown in Examples, and also have hygroscopic characteristics as shown in the data of Test Examples described later. It can be used as a drug substance for pharmaceuticals.

図1は、実施例1で得られた化合物(I)・1.5コハク酸塩の結晶の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。1 is a powder X-ray diffraction pattern of the crystal of compound (I) · 1.5 succinate obtained in Example 1. FIG. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図2は、実施例2で得られた化合物(I)・0.5コハク酸塩の結晶(α)の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 2 is a powder X-ray diffraction pattern of the crystal (α) of compound (I) · 0.5 succinate obtained in Example 2. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図3は、実施例3で得られた化合物(I)・マレイン酸塩の結晶の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 3 is a powder X-ray diffraction pattern of the crystal of compound (I) · maleate obtained in Example 3. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図4は、実施例1で得られた化合物(I)・1.5コハク酸塩の結晶の13C固体NMRスペクトルである。4 is a 13C solid state NMR spectrum of the crystal of compound (I) · 1.5 succinate obtained in Example 1. FIG. 図5は、実施例1で得られた化合物(I)・1.5コハク酸塩の結晶の吸湿性を示したグラフである。横軸は相対湿度、縦軸は重量変化を示す。FIG. 5 is a graph showing the hygroscopicity of the crystals of compound (I) · 1.5 succinate obtained in Example 1. The horizontal axis represents relative humidity, and the vertical axis represents weight change. 図6は、製造例1−15で得られた化合物(I)・フリー体の結晶(Free Form A)の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 6 is a powder X-ray diffraction pattern of the compound (I) · free form crystal (Free Form A) obtained in Production Example 1-15. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図7は、参考例1で得られた化合物(I)・フリー体の結晶(Free Form B)の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 7 is a powder X-ray diffraction pattern of the compound (I) · free form crystal (Free Form B) obtained in Reference Example 1. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図8は、参考例2で得られた化合物(I)・フリー体の結晶(Free Form Hydrate)の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 8 is a powder X-ray diffraction pattern of the compound (I) · free form crystal obtained in Reference Example 2 (Free Form Hydrate). The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図9は、参考例3で得られた化合物(I)・メシル酸塩の結晶の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 9 is a powder X-ray diffraction pattern of the compound (I) mesylate crystal obtained in Reference Example 3. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図10は、参考例4で得られた化合物(I)・トシル酸塩の結晶の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 10 is a powder X-ray diffraction pattern of the compound (I) -tosylate crystal obtained in Reference Example 4. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図11は、参考例5で得られた化合物(I)・安息香酸塩の結晶の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 11 is a powder X-ray diffraction pattern of crystals of compound (I) .benzoate obtained in Reference Example 5. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図12は、参考例6で得られた化合物(I)・フマル酸塩の結晶の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 12 is a powder X-ray diffraction pattern of the crystal of compound (I) • fumarate obtained in Reference Example 6. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図13は、実施例4で得られた化合物(I)・0.5コハク酸塩の結晶(α)の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 13 is a powder X-ray diffraction pattern of the crystal (α) of compound (I) · 0.5 succinate obtained in Example 4. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図14は、実施例5で得られた化合物(I)・0.5コハク酸塩の結晶(β)の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。14 is a powder X-ray diffraction pattern of compound (I) · 0.5 succinate crystal (β) obtained in Example 5. FIG. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity. 図15は、実施例6で得られた化合物(I)・1.5コハク酸塩の非晶質の粉末X線回折パターンである。横軸は回折角(2θ)、縦軸はピーク強度を示す。FIG. 15 is an amorphous powder X-ray diffraction pattern of the compound (I) · 1.5 succinate obtained in Example 6. The horizontal axis indicates the diffraction angle (2θ), and the vertical axis indicates the peak intensity.

次に、本発明の化合物(I)の塩およびその結晶、ならびにそれらの製造方法について詳細に説明する。   Next, the salt of the compound (I) of the present invention, the crystal thereof, and the production method thereof will be described in detail.

本明細書において、「塩」とは、塩基性成分である化合物(I)と、化合物(I)に対して特定の当量数の酸とからなる化学物質を意味する。   In the present specification, the “salt” means a chemical substance composed of the basic component compound (I) and a specific equivalent number of acids relative to the compound (I).

本明細書において使用する「塩」としては、例えば、無機酸との塩、有機酸との塩、酸性アミノ酸との塩等が挙げられ、中でも薬剤学的に許容される塩が好ましい。   Examples of the “salt” used in the present specification include a salt with an inorganic acid, a salt with an organic acid, a salt with an acidic amino acid, and the like. Among them, a pharmaceutically acceptable salt is preferable.

無機酸との塩の例としては、例えば、塩酸、臭化水素酸、硫酸、硝酸、リン酸等との塩が挙げられ、有機酸との塩の例としては、例えば酢酸、コハク酸、フマル酸、マレイン酸、酒石酸、リンゴ酸、クエン酸、乳酸、ステアリン酸、安息香酸等の有機カルボン酸との塩、メタンスルホン酸(メシル酸)、エタンスルホン酸、ベンゼンスルホン酸、p−トルエンスルホン酸(トシル酸)等の有機スルホン酸との塩が挙げられ、中でも、コハク酸、マレイン酸が好ましく、コハク酸が特に好ましい。   Examples of salts with inorganic acids include, for example, salts with hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, etc. Examples of salts with organic acids include, for example, acetic acid, succinic acid, fumaric acid. Salts with organic carboxylic acids such as acid, maleic acid, tartaric acid, malic acid, citric acid, lactic acid, stearic acid, benzoic acid, methanesulfonic acid (mesyl acid), ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid Examples thereof include salts with organic sulfonic acids such as (tosylic acid), among which succinic acid and maleic acid are preferable, and succinic acid is particularly preferable.

酸性アミノ酸との塩の例としては、例えばアスパラギン酸、グルタミン酸等との塩が挙げられる。   Examples of salts with acidic amino acids include salts with aspartic acid, glutamic acid and the like.

本発明の塩は、無水物または、水和物もしくは溶媒和物でもよい。本明細書において、水和物または溶媒和物とは、化合物(I)またはその塩と、水分子または溶媒分子とがそれぞれ一緒になって形成する固体をいい、その固体は結晶であってもよく、溶媒和物の溶媒としては、例えば、アセトン、2−ブタノン、シクロヘキサノンなどのケトン系溶媒;酢酸メチル、酢酸エチル等のエステル系溶媒;1,2−ジメトキシエタン、t−ブチルメチルエーテルのようなエーテル系溶媒;メタノール、エタノール、1−プロパノール、イソプロパノール等のアルコール系溶媒、N−メチル−2−ピロリドン、N,N−ジメチルホルムアミド、ジメチルスルホキシド等の極性溶媒が挙げられる。化合物(I)またはその塩に対する水分子または溶媒分子の数は特に限定されず、例えば、1分子または2分子であってもよい。   The salt of the present invention may be an anhydride, hydrate or solvate. In this specification, the hydrate or solvate refers to a solid formed by combining Compound (I) or a salt thereof with a water molecule or a solvent molecule, respectively, and the solid may be a crystal. Well, as a solvent of the solvate, for example, ketone solvents such as acetone, 2-butanone and cyclohexanone; ester solvents such as methyl acetate and ethyl acetate; 1,2-dimethoxyethane and t-butyl methyl ether And ether solvents; alcohol solvents such as methanol, ethanol, 1-propanol and isopropanol; polar solvents such as N-methyl-2-pyrrolidone, N, N-dimethylformamide and dimethyl sulfoxide. The number of water molecules or solvent molecules for compound (I) or a salt thereof is not particularly limited, and may be one molecule or two molecules, for example.

本明細書において、「結晶」とは、化合物(I)またはその塩の結晶を指す。したがって、例えば化合物(I)・1.5コハク酸塩の結晶とは、化合物(I)とコハク酸とで形成される塩の結晶であって、化合物(I)1分子に対してコハク酸を1.5分子含有する塩の結晶を意味する。   In the present specification, “crystal” refers to a crystal of compound (I) or a salt thereof. Thus, for example, a crystal of compound (I) · 1.5 succinate is a crystal of a salt formed from compound (I) and succinic acid, and succinic acid is added to one molecule of compound (I). It means a salt crystal containing 1.5 molecules.

本明細書において、好ましい結晶としては、粉末X線回折において、
回折角度(2θ±0.2°)22.4°に回折ピークを有する、化合物(I)・1.5コハク酸塩の結晶;
回折角度(2θ±0.2°)22.4°および25.3°に回折ピークを有する、化合物(I)・1.5コハク酸塩の結晶;
回折角度(2θ±0.2°)22.4°、25.3°および23.3°に回折ピークを有する、化合物(I)・1.5コハク酸塩の結晶;
回折角度(2θ±0.2°)22.4°、25.3°、23.3°、13.2°および22.0°に回折ピークを有する、化合物(I)・1.5コハク酸塩の結晶;
回折角度(2θ±0.2°)22.4°、25.3°、23.3°、13.2°、22.0°、19.3°、15.7°、22.7°、20.6°および16.0°に回折ピークを有する、化合物(I)・1.5コハク酸塩の結晶;
回折角度(2θ±0.2°)19.8°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(α);
回折角度(2θ±0.2°)19.8°および15.7°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(α);
回折角度(2θ±0.2°)19.8°、15.7°および13.9°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(α);
回折角度(2θ±0.2°)19.8°、15.7°、13.9°、21.4°および25.0°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(α);
回折角度(2θ±0.2°)19.8°、15.7°、13.9°、21.4°、25.0°、20.6°、18.2°、26.8°、18.8°および22.4°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(α);
回折角度(2θ±0.2°)16.6°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(β);
回折角度(2θ±0.2°)16.6°および19.7°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(β);
回折角度(2θ±0.2°)16.6°、19.7°および15.7°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(β);
回折角度(2θ±0.2°)16.6°、19.7°、15.7°、9.3°および14.3°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(β);
回折角度(2θ±0.2°)16.6°、19.7°、15.7°、9.3°、14.3°、21.8°、20.6°、18.7°、18.1°および26.5°に回折ピークを有する、化合物(I)・0.5コハク酸塩の結晶(β);
回折角度(2θ±0.2°)20.1°に回折ピークを有する、化合物(I)・マレイン酸塩の結晶;
回折角度(2θ±0.2°)20.1°および17.0°に回折ピークを有する、化合物(I)・マレイン酸塩の結晶;
回折角度(2θ±0.2°)20.1°、17.0°および16.2°に回折ピークを有する、化合物(I)・マレイン酸塩の結晶;
回折角度(2θ±0.2°)20.1°、17.0°、16.2°、22.8°および21.9°に回折ピークを有する、化合物(I)・マレイン酸塩の結晶;
回折角度(2θ±0.2°)20.1°、17.0°、16.2°、22.8°、21.9°、25.8°、9.0°、15.2°、24.3°および19.6°に回折ピークを有する、化合物(I)・マレイン酸塩の結晶;
13C固体NMRスペクトルにおいて、化学シフト(±0.5ppm)108.5ppm、155.1ppmおよび179.9ppmにピークを有する、化合物(I)・1.5コハク酸塩の結晶;
または、
13C固体NMRスペクトルにおいて、化学シフト(±0.5ppm)27.1ppm、34.8ppm、108.5ppm、155.1ppmおよび179.9ppmにピークを有する、化合物(I)・1.5コハク酸塩の結晶などを挙げることができる。
In this specification, as a preferable crystal, in powder X-ray diffraction,
Compound (I) 1.5 succinate crystals having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 22.4 °;
Crystals of compound (I) · 1.5 succinate having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 22.4 ° and 25.3 °;
Crystals of compound (I) 1.5 succinate having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 22.4 °, 25.3 ° and 23.3 °;
Compound (I) 1.5 succinic acid having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 22.4 °, 25.3 °, 23.3 °, 13.2 ° and 22.0 ° Salt crystals;
Diffraction angle (2θ ± 0.2 °) 22.4 °, 25.3 °, 23.3 °, 13.2 °, 22.0 °, 19.3 °, 15.7 °, 22.7 °, Crystals of compound (I) · 1.5 succinate having diffraction peaks at 20.6 ° and 16.0 °;
Crystal (α) of compound (I) 0.5 succinate having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 19.8 °;
Crystals of compound (I) 0.5 succinate (α) having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 19.8 ° and 15.7 °;
Crystals of compound (I) 0.5 succinate (α) having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 19.8 °, 15.7 ° and 13.9 °;
Compound (I) 0.5 succinic acid having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 19.8 °, 15.7 °, 13.9 °, 21.4 ° and 25.0 ° Salt crystals (α);
Diffraction angle (2θ ± 0.2 °) 19.8 °, 15.7 °, 13.9 °, 21.4 °, 25.0 °, 20.6 °, 18.2 °, 26.8 °, Compound (I) 0.5 succinate crystals (α) having diffraction peaks at 18.8 ° and 22.4 °;
Compound (I) -0.5 succinate crystal (β) having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 16.6 °;
Compound (I) -0.5 succinate crystal (β) having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 16.6 ° and 19.7 °;
Crystals of compound (I) and 0.5 succinate (β) having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 16.6 °, 19.7 ° and 15.7 °;
Compound (I) 0.5 succinic acid having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 16.6 °, 19.7 °, 15.7 °, 9.3 ° and 14.3 ° Salt crystals (β);
Diffraction angle (2θ ± 0.2 °) 16.6 °, 19.7 °, 15.7 °, 9.3 °, 14.3 °, 21.8 °, 20.6 °, 18.7 °, Crystals (β) of compound (I) 0.5 succinate having diffraction peaks at 18.1 ° and 26.5 °;
Compound (I) maleate crystal having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 20.1 °;
Compound (I) maleate crystals having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 20.1 ° and 17.0 °;
Crystals of Compound (I) maleate having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 20.1 °, 17.0 ° and 16.2 °;
Crystals of compound (I) maleate having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 20.1 °, 17.0 °, 16.2 °, 22.8 ° and 21.9 ° ;
Diffraction angle (2θ ± 0.2 °) 20.1 °, 17.0 °, 16.2 °, 22.8 °, 21.9 °, 25.8 °, 9.0 °, 15.2 °, Compound (I) maleate crystals having diffraction peaks at 24.3 ° and 19.6 °;
Compound (I) -1.5 succinate crystals having peaks at chemical shifts (± 0.5 ppm) 108.5 ppm, 155.1 ppm and 179.9 ppm in the 13 C solid state NMR spectrum;
Or
Compound (I) 1.5 succinate having peaks at chemical shift (± 0.5 ppm) 27.1 ppm, 34.8 ppm, 108.5 ppm, 155.1 ppm and 179.9 ppm in 13 C solid state NMR spectrum And the like.

上記記載の粉末X線回折における回折ピークは、化合物(I)・1.5コハク酸塩の結晶、化合物(I)・0.5コハク酸塩の結晶(α)と(β)、化合物(I)・マレイン酸塩の結晶にそれぞれ特有なものであり、当該結晶に特徴的な回折ピークである。   The diffraction peaks in the powder X-ray diffraction described above are: compound (I) · 1.5 succinate crystal, compound (I) · 0.5 succinate crystal (α) and (β), compound (I) )-It is unique to each maleate crystal and is a diffraction peak characteristic of the crystal.

一般に、粉末X線回折における回折角度(2θ)は±0.2°の範囲内で誤差が生じ得るため、上記の回折角度の値は±0.2°程度の範囲内の数値も含むものとして理解される必要がある。したがって、特定の塩において、粉末X線回折におけるピークの回折角度が完全に一致する結晶だけでなく、ピークの回折角度が±0.2°程度の誤差で一致する結晶も同一であり、本発明に含まれる。   In general, since the diffraction angle (2θ) in powder X-ray diffraction may cause an error within a range of ± 0.2 °, the above diffraction angle value includes a numerical value within a range of about ± 0.2 °. Need to be understood. Therefore, in a specific salt, not only a crystal in which the diffraction angle of a peak in powder X-ray diffraction completely matches, but also a crystal in which the diffraction angle of a peak matches with an error of about ± 0.2 ° is the same. include.

本明細書において、例えば、「回折角度(2θ±0.2°)22.4°に回折ピークを有する」とは、「回折角度(2θ)22.2°〜22.6°に回折ピークを有する」ということを意味し、その他の回折角度の場合も同様である。   In this specification, for example, “having a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 22.4 °” means “having a diffraction peak at a diffraction angle (2θ) of 22.2 ° to 22.6 °. The same applies to other diffraction angles.

また、一般に、粉末X線回折における回折角度(2θ)のピーク強度または半値幅は、結晶形が同一であっても、測定条件の違いや測定試料として用いる粉末結晶の各粒子の大きさや形状のばらつきにより、測定ごとに異なり、必ずしも一定のピーク強度または半値幅が常に示されるとは限らない。そのため、粉末X線回折パターンの比較において、同じ回折角度(2θ)で、そのピーク強度または半値幅に違いがあっても、その違いは、異なる結晶形に由来することを意味するものではない。したがって、本発明の特定の塩の結晶に特徴的な回折ピークに対して、そのような違いを有する粉末X線回折パターンの当該塩の結晶は、本発明の塩の結晶と同一の結晶形であることを意味する。また、本明細書において「図1の粉末X線回折パターンを有する」とは、特徴的な回折ピークを有する粉末X線回折パターンが、図1で示されるパターンに完全に一致する場合だけでなく、特徴的な回折角度が一致するもののピーク強度または半値幅が異なる粉末X線回折パターンの場合であっても、図1で示される粉末X線回折パターンを有することを意味する。したがって、このような粉末X線回折パターンを有する結晶は全て、本発明の結晶と同一の結晶であることを意味する。   In general, the peak intensity or half-value width of the diffraction angle (2θ) in powder X-ray diffraction is the difference in measurement conditions and the size and shape of each particle of a powder crystal used as a measurement sample even if the crystal form is the same. Due to variations, it varies from measurement to measurement, and a constant peak intensity or full width at half maximum is not always indicated. Therefore, in the comparison of powder X-ray diffraction patterns, even if there is a difference in peak intensity or half-value width at the same diffraction angle (2θ), the difference does not mean that the difference is derived from different crystal forms. Therefore, the crystal of the salt of the powder X-ray diffraction pattern having such a difference with respect to the diffraction peak characteristic of the crystal of the specific salt of the present invention has the same crystal form as the crystal of the salt of the present invention. It means that there is. Further, in the present specification, “having the powder X-ray diffraction pattern of FIG. 1” means not only the case where the powder X-ray diffraction pattern having a characteristic diffraction peak completely matches the pattern shown in FIG. This means that the powder X-ray diffraction pattern shown in FIG. 1 is obtained even in the case of a powder X-ray diffraction pattern having the same diffraction angle but different peak intensity or half width. Therefore, it means that all crystals having such a powder X-ray diffraction pattern are the same crystals as the crystal of the present invention.

本明細書において、「化学シフト(±0.5ppm)27.1ppm、34.8ppm、108.5ppm、155.1ppmおよび179.9ppm」とは、「通常の測定条件または本明細書と実質的に同一の条件にて13C固体NMRスペクトル測定を行い、それぞれ、化学シフト(±0.5ppm)27.1ppm、34.8ppm、108.5ppm、155.1ppmおよび179.9ppmと実質的に同等なピークを有すること」を意味する。In this specification, “chemical shift (± 0.5 ppm) 27.1 ppm, 34.8 ppm, 108.5 ppm, 155.1 ppm and 179.9 ppm” means “normal measurement conditions or substantially the same as the present specification. 13 C solid state NMR spectra were measured under the same conditions, and peaks substantially equivalent to chemical shifts (± 0.5 ppm) of 27.1 ppm, 34.8 ppm, 108.5 ppm, 155.1 ppm and 179.9 ppm, respectively. It means "having".

「実質的に同等なピークを有する」か否かの判断に際して、一般に、13C固体NMRスペクトルにおける化学シフト(ppm)は、±0.5ppmの範囲内で誤差が生じ得るため、上記の化学シフトの値は、±0.5ppm程度の範囲内の数値も含むものとして理解される必要がある。したがって、13C固体NMRスペクトルにおける化学シフトが完全に一致する結晶だけでなく、化学シフトが±0.5ppm程度の誤差で一致する結晶も本発明に含まれる。それ故に、本明細書において、例えば、「化学シフト(±0.5ppm)27.1ppmにピークを有する」とは、「化学シフト26.6ppm〜27.6ppmにピークを有する」ことを意味し、その他の13C固体NMRスペクトルにおける化学シフトの場合も同様である。In determining whether or not it has “substantially equivalent peaks”, in general, the chemical shift (ppm) in a 13 C solid state NMR spectrum can cause an error within a range of ± 0.5 ppm. The value of should be understood as including values in the range of about ± 0.5 ppm. Therefore, the present invention includes not only crystals in which the chemical shift in the 13 C solid state NMR spectrum is completely matched but also crystals in which the chemical shift is matched with an error of about ± 0.5 ppm. Therefore, in the present specification, for example, “having a peak at 27.1 ppm of chemical shift (± 0.5 ppm)” means “having a peak at 26.6 ppm to 27.6 ppm of chemical shift” The same applies to chemical shifts in other 13 C solid state NMR spectra.

以下に、本発明の一実施形態である化合物(I)の塩、結晶等の製造方法について説明する。   Below, the manufacturing method of the salt of the compound (I) and crystal | crystallization which are one Embodiment of this invention is demonstrated.

化合物(I)の製造
化合物(I)は、後述する製造例1に記載の方法で合成することができる。
Production of Compound (I) Compound (I) can be synthesized by the method described in Production Example 1 described later.

化合物(I)の塩の製造方法
本発明に係る化合物(I)の塩は、通常の塩を製造する方法により得ることができる。具体的には、例えば、化合物(I)を溶媒に、必要に応じて加温して、懸濁または溶解させ、次いで、得られる懸濁液または溶液に、酸を加え、室温下あるいは冷却しながら数分から数日間、撹拌または放置することにより、製造することができる。これらの製造方法により、化合物(I)の塩を、結晶または非晶質として得ることができる。また、非晶質は、これらの製造方法に、必要に応じて、さらに凍結乾燥等の操作を行うことにより得ることもできる。ここで使用する溶媒としては、例えばエタノール、1−プロパノール、イソプロパノール等のアルコール系溶媒;アセトニトリル;アセトン、2−ブタノン等のケトン系溶媒;酢酸エチル等のエステル系溶媒;ヘキサン、ヘプタン等の飽和炭化水素系溶媒;t−ブチルメチルエーテル等のエーテル系溶媒または水を挙げることができる。これらの溶媒は単独で使用してもよく、2種以上を混合して使用してもよい。
Method for Producing Compound (I) Salt The salt of compound (I) according to the present invention can be obtained by a method for producing an ordinary salt. Specifically, for example, compound (I) is heated or suspended in a solvent as necessary to suspend or dissolve it, and then an acid is added to the resulting suspension or solution and cooled at room temperature or cooled. However, it can be produced by stirring or standing for several minutes to several days. By these production methods, the salt of the compound (I) can be obtained as a crystal or amorphous. Amorphous can also be obtained by subjecting these production methods to operations such as freeze-drying as necessary. Examples of the solvent used here include alcohol solvents such as ethanol, 1-propanol and isopropanol; acetonitrile; ketone solvents such as acetone and 2-butanone; ester solvents such as ethyl acetate; saturated carbonization such as hexane and heptane. Examples of the hydrogen solvent include ether solvents such as t-butyl methyl ether or water. These solvents may be used alone or in combination of two or more.

化合物(I)またはその塩の結晶の製造方法
化合物(I)またはその塩の結晶は、上述の化合物(I)の製造方法、またはその塩の製造方法により製造することができ、または、化合物(I)またはその塩を、溶媒中で加熱溶解し、攪拌下冷却して晶析することにより、製造することもできる。
Method for Producing Crystal of Compound (I) or a Salt thereof Crystal of Compound (I) or a salt thereof can be produced by the above-described method for producing Compound (I) or a method for producing a salt thereof, or Compound (I) It can also be produced by dissolving I) or a salt thereof in a solvent by heating, cooling with stirring and crystallization.

晶析に使用する化合物(I)またはその塩は、どのような形態であってもよく、溶媒和物もしくは水和物または無水物でもよく、非晶質でも結晶質(複数の結晶多形からなるものを含む)でもよく、これらの混合物でもよい。   Compound (I) or a salt thereof used for crystallization may be in any form, and may be a solvate, hydrate, or anhydride, and may be amorphous or crystalline (from a plurality of crystal polymorphs). Or a mixture thereof.

晶析に使用する溶媒は、例えば、メタノール、エタノール、イソプロパノール、1−プロパノール等のアルコール系溶媒;アセトニトリル;N,N−ジメチルホルムアミド等のアミド系溶媒;酢酸エチル等のエステル系溶媒;ヘキサン、ヘプタン等の飽和炭化水素系溶媒;アセトン、2−ブタノン等のケトン系溶媒;t−ブチルメチルエーテル等のエーテル系溶媒または水を挙げることができる。また、これらの溶媒は単独で使用してもよく、2種以上を混合して使用してもよい。   Solvents used for crystallization are, for example, alcohol solvents such as methanol, ethanol, isopropanol and 1-propanol; acetonitrile; amide solvents such as N, N-dimethylformamide; ester solvents such as ethyl acetate; hexane and heptane Saturated hydrocarbon solvents such as acetone; ketone solvents such as acetone and 2-butanone; ether solvents such as t-butyl methyl ether or water. These solvents may be used alone or in combination of two or more.

溶媒の使用量は、化合物(I)またはその塩が加熱により溶解する量または懸濁液が撹拌可能となる量を下限とし、結晶の収量が著しく低下しない量を上限として適宜選択することができる。   The amount of the solvent used can be appropriately selected with the lower limit being the amount in which the compound (I) or a salt thereof is dissolved by heating or the amount allowing the suspension to be stirred, and the upper limit being the amount that does not significantly reduce the yield of crystals. .

晶析において、種結晶(所望の化合物(I)またはその塩の結晶など)を加えても、加えなくてもよい。種結晶を加える温度は、特に限定されないが、好ましくは0〜80℃である。   In crystallization, seed crystals (such as crystals of desired compound (I) or a salt thereof) may or may not be added. Although the temperature which adds a seed crystal is not specifically limited, Preferably it is 0-80 degreeC.

化合物(I)またはその塩を加熱して溶解する場合の温度は、溶媒に応じて化合物(I)またはその塩が溶解する温度を適宜選択すればよいが、好ましくは50℃から再結晶溶媒が還流を開始する温度の範囲であり、より好ましくは55〜80℃である。   The temperature at which compound (I) or a salt thereof is dissolved by heating may be appropriately selected according to the solvent, and the temperature at which compound (I) or a salt thereof is dissolved is preferably selected. It is the range of the temperature which starts reflux, More preferably, it is 55-80 degreeC.

晶析時の冷却は、急冷すると態様の異なる結晶(多形)を含むものを与えうるので、結晶の品質や粒度等への影響を考慮して適宜冷却速度を調整して実施することが望ましく、好ましくは、例えば5〜40℃/時間の速度での冷却である。より好ましくは、例えば5〜25℃/時間の速度での冷却である。   Cooling during crystallization can give crystals containing different forms of crystals (polymorphs) when rapidly cooled, so it is desirable to adjust the cooling rate appropriately in consideration of the effects on crystal quality and particle size. Preferably, cooling is performed at a rate of 5 to 40 ° C./hour, for example. More preferably, it is cooling at a rate of, for example, 5 to 25 ° C./hour.

また、最終的な晶析温度は、結晶の収量と品質等から適宜選択することができるが、好ましくは−25〜30℃である。   The final crystallization temperature can be appropriately selected from the yield and quality of crystals, but is preferably −25 to 30 ° C.

晶析した結晶を通常の濾過操作で分離し、必要に応じてろ別した結晶を溶媒で洗浄し、さらにこれを乾燥して、目的の結晶を得ることができる。結晶の洗浄に使用する溶媒には、晶析溶媒と同様のものを使用できる。好ましくは、例えば、エタノール、アセトン、2−ブタノン、酢酸エチル、ジエチルエーテル、t−ブチルメチルエーテル、ヘキサン等を挙げることができる。また、これらの溶媒は単独で使用してもよく、2種以上を混合して使用してもよい。   The crystallized crystal is separated by a normal filtration operation, and the crystal separated by filtration as necessary is washed with a solvent and further dried to obtain the desired crystal. As the solvent used for washing the crystals, the same crystallization solvent can be used. Preferable examples include ethanol, acetone, 2-butanone, ethyl acetate, diethyl ether, t-butyl methyl ether, and hexane. These solvents may be used alone or in combination of two or more.

濾過操作で分離した結晶は、適宜、大気下または窒素気流下に放置することにより、または加熱によって乾燥することができる。   The crystals separated by the filtration operation can be appropriately dried by being left in the air or a nitrogen stream or by heating.

乾燥時間は、残留溶媒が所定の量を下回るまでの時間を製造量、乾燥装置、乾燥温度等に応じて適宜選択すればよい。また、乾燥は通風下でも減圧下でも行うことができる。減圧度は、製造量、乾燥装置、乾燥温度等に応じて適宜選択すればよい。得られた結晶は、乾燥後、必要に応じて大気中に放置することもできる。   The drying time may be appropriately selected according to the production amount, drying apparatus, drying temperature, etc. until the residual solvent falls below a predetermined amount. Moreover, drying can be performed under ventilation or under reduced pressure. The degree of vacuum may be appropriately selected according to the production amount, the drying device, the drying temperature, and the like. The obtained crystals can be left in the air after drying, if necessary.

化合物(I)の塩およびその結晶は、通常の方法により製剤化が可能であり、剤形としては、例えば、経口剤(錠剤、顆粒剤、散剤、カプセル剤、シロップ剤等)、注射剤(静脈内投与用、筋肉内投与用、皮下投与用、腹腔内投与用等)、外用剤(経皮吸収製剤(軟膏剤、貼付剤等)、点眼剤、点鼻剤、坐剤等)とすることができる。   The salt of Compound (I) and crystals thereof can be formulated by a conventional method. Examples of the dosage form include oral preparations (tablets, granules, powders, capsules, syrups, etc.), injections ( For intravenous administration, intramuscular administration, subcutaneous administration, intraperitoneal administration, etc.), external preparations (transdermal absorption preparations (ointments, patches, etc.), eye drops, nasal drops, suppositories, etc.) be able to.

経口用固形製剤を製造する場合には、化合物(I)の塩またはその結晶に、必要に応じて、賦形剤、結合剤、崩壊剤、滑沢剤、着色剤等を添加し、常法により錠剤、顆粒剤、散剤、カプセル剤を製造することができる。また、錠剤、顆粒剤、散剤、カプセル剤等は、必要に応じて、コーティングを施してもよい。
賦形剤としては、例えば、乳糖、結晶セルロース等を、結合剤としては、例えば、ヒドロキシプロピルセルロース等を、崩壊剤としては、例えば、クロスカルメロースナトリウム等を、滑沢剤としては、例えば、ステアリン酸マグネシウム等を、着色剤としては、例えば、酸化チタン等を、コーティング剤としては、例えば、ヒドロキシプロピルメチルセルロース等を挙げることができるが、これらに限定されるものではない。
これらの錠剤、カプセル剤、顆粒剤、散剤等の固形製剤は、通常、医薬品として利用できる薬効を示す限り、任意の量の化合物(I)の塩またはその結晶を含むことができる。
When producing an oral solid preparation, an excipient, a binder, a disintegrant, a lubricant, a coloring agent, etc. are added to the salt of compound (I) or a crystal thereof as necessary. Thus, tablets, granules, powders and capsules can be produced. Tablets, granules, powders, capsules and the like may be coated as necessary.
As the excipient, for example, lactose, crystalline cellulose and the like, as the binder, for example, hydroxypropyl cellulose and the like, as the disintegrant, for example, croscarmellose sodium and the like, as the lubricant, for example, Examples of the colorant include magnesium stearate, titanium oxide, and the like, and examples of the coating agent include, but are not limited to, hydroxypropylmethylcellulose and the like.
These solid preparations such as tablets, capsules, granules, powders and the like can usually contain any amount of the salt of compound (I) or a crystal thereof as long as it shows a medicinal effect that can be used as a medicine.

注射剤(静脈内投与用、筋肉内投与用、皮下投与用、腹腔内投与用等)を製造する場合には、化合物(I)の塩またはその結晶に、必要に応じて、pH調整剤、緩衝剤、懸濁化剤、溶解補助剤、抗酸化剤、保存剤(防腐剤)、等張化剤等を添加し、常法により注射剤を製造することができる。また、凍結乾燥して、用時溶解型の凍結乾燥製剤としてもよい。
pH調整剤や緩衝剤としては、例えば、有機酸または無機酸および/またはその薬剤学的に許容される塩等を、懸濁化剤としては、例えば、ヒドロキシプロピルセルロース等を、溶解補助剤としては、例えば、ポリソルベート80等を、抗酸化剤としては、例えば、α−トコフェロール等を、保存剤としては、例えば、パラオキシ安息香酸メチル、パラオキシ安息香酸エチル等を、等張化剤としては、例えば、ブドウ糖等を挙げることができるが、これらに限定されるものではない。
これらの注射剤は、通常、医薬品として利用できる薬効を示す限り、任意の量の化合物(I)の塩またはその結晶を含むことができる。
In the case of producing an injection (for intravenous administration, intramuscular administration, subcutaneous administration, intraperitoneal administration, etc.), if necessary, a pH adjusting agent is added to the salt of compound (I) or a crystal thereof. Buffering agents, suspending agents, solubilizing agents, antioxidants, preservatives (preservatives), isotonic agents and the like can be added to produce injections by conventional methods. Alternatively, it may be freeze-dried to obtain a freeze-dried preparation that is dissolved at the time of use.
Examples of pH adjusters and buffers include organic acids or inorganic acids and / or pharmaceutically acceptable salts thereof, and examples of suspending agents include hydroxypropyl cellulose and the like as solubilizers. Is, for example, polysorbate 80, etc., as an antioxidant, for example, α-tocopherol, etc., as a preservative, for example, methyl paraoxybenzoate, ethyl paraoxybenzoate, etc. , Glucose and the like can be mentioned, but are not limited thereto.
These injections can usually contain any amount of the salt of compound (I) or a crystal thereof as long as it shows a medicinal effect that can be used as a medicine.

外用剤を製造する場合には、化合物(I)の塩またはその結晶に、基剤原料を添加し、必要に応じて、例えば、前記の保存剤、pH調整剤、抗酸化剤、着色剤等を加えて、常法により、例えば、経皮吸収製剤(軟膏剤、貼付剤等)、点眼剤、点鼻剤、坐剤等を製造することができる。
使用する基剤原料としては、例えば、医薬品、医薬部外品、化粧品等に通常使用される各種原料を用いることが可能である。具体的には例えば、動植物油、鉱物油、エステル油、ワックス類、乳化剤、高級アルコール類、脂肪酸類、シリコン油、界面活性剤、リン脂質類、アルコール類、多価アルコール類、水溶性高分子類、粘土鉱物類、精製水等の原料を挙げることができる。
これらの外用剤は、通常、医薬品として利用できる薬効を示す限り、任意の量の化合物(I)の塩またはその結晶を含むことができる。
In the case of producing an external preparation, a base material is added to the salt of compound (I) or a crystal thereof, and if necessary, for example, the above-mentioned preservative, pH adjuster, antioxidant, coloring agent, etc. In addition, for example, transdermal preparations (ointments, patches, etc.), eye drops, nasal drops, suppositories and the like can be produced by conventional methods.
As the base material to be used, for example, various raw materials usually used for pharmaceuticals, quasi drugs, cosmetics and the like can be used. Specifically, for example, animal and vegetable oils, mineral oils, ester oils, waxes, emulsifiers, higher alcohols, fatty acids, silicone oils, surfactants, phospholipids, alcohols, polyhydric alcohols, water-soluble polymers And raw materials such as clay minerals and purified water.
These external preparations can usually contain any amount of a salt of compound (I) or a crystal thereof as long as it shows a medicinal effect that can be used as a pharmaceutical product.

化合物(I)の塩またはその結晶の投与量は、症状の程度、年齢、性別、体重、投与形態・塩の種類、疾患の具体的な種類等に応じて異なり、許容できない副作用を引き起こすことなく投与できる薬物の最大の用量を超えなければ限定されないが、通常、成人の場合は1日あたり経口投与で約30μg〜10g、例えば100μg〜5g、さらに例えば100μg〜1gを、注射投与で約30μg〜1g、例えば100μg〜500mg、さらに例えば100μg〜300mgをそれぞれ1回または数回に分けて投与する。   The dosage of the salt of Compound (I) or crystals thereof varies depending on the degree of symptoms, age, sex, body weight, dosage form / salt type, specific type of disease, etc., without causing unacceptable side effects. Although it is not limited as long as the maximum dose of the drug that can be administered is not exceeded, it is usually about 30 μg to 10 g, such as 100 μg to 5 g, for example 100 μg to 1 g, or about 30 μg 1 g, for example, 100 μg to 500 mg, further 100 μg to 300 mg, for example, is administered once or in several divided doses.

本発明に係る化合物は、例えば以下の製造例および実施例に記載した方法により製造することができる。ただし、これらは例示的なものであって、本発明に係る化合物はいかなる場合も以下の具体例に限定されるものではない。   The compounds according to the present invention can be produced, for example, by the methods described in the following production examples and examples. However, these are illustrative, and the compound according to the present invention is not limited to the following specific examples in any case.

以下の製造例、実施例および参考例において製造された結晶の粉末X線結晶回折は、得られた結晶を、粉末X線回折装置の試料台に載せ、下記の条件で分析した。その結果を図1〜3および6〜15に示す。   In the powder X-ray crystal diffraction of the crystals produced in the following production examples, examples and reference examples, the obtained crystals were placed on a sample stage of a powder X-ray diffractometer and analyzed under the following conditions. The results are shown in FIGS. 1-3 and 6-15.

測定条件
サンプルホルダー:アルミニウム
ターゲット:銅
検出器:シンチレーションカウンター
管電圧:50kV
管電流:300mA
スリット:DS 0.5 mm(Height limiting slit 2 mm),SS Open, RS Open
走査速度:10°/分
サンプリング間隔:0.02°
走査範囲:5〜35°
Measurement conditions Sample holder: Aluminum Target: Copper Detector: Scintillation counter Tube voltage: 50kV
Tube current: 300mA
Slit: DS 0.5 mm (Height limiting slit 2 mm), SS Open, RS Open
Scanning speed: 10 ° / min Sampling interval: 0.02 °
Scanning range: 5-35 °

結晶の13C固体NMRスペクトルは以下の条件で測定した。その結果を図4に示す。
測定条件
使用装置:AVANCE400(BRUKER社製)
測定温度:室温(22℃)
基準物質:グリシン(外部基準:176.03ppm)
測定核:13C(100.6248425MHz)
パルス繰り返し時間:3秒
パルスモード:TOSS測定
The 13 C solid state NMR spectrum of the crystal was measured under the following conditions. The result is shown in FIG.
Measuring condition use device: AVANCE400 (manufactured by BRUKER)
Measurement temperature: Room temperature (22 ° C)
Reference substance: Glycine (External standard: 176.03 ppm)
Measurement nucleus: 13C (100.6248425MHz)
Pulse repetition time: 3 seconds Pulse mode: TOSS measurement

製造例中、特に記載がない場合は、シリカゲルカラムクロマトグラフィーに使用している精製用シリカゲルとしては、Silica gel 60(Kanto Chemicals)またはPresep Silica Gel(WAKO)を用いた。また、NHシリカゲルカラムクロマトグラフィーに使用している精製用シリカゲルは、NHシリカゲル(Fuji Silysia Chemical LTD.)またはHi−Flash Column Amino(YAMAZENE CORPORATION)を用いた。   In the production examples, unless otherwise specified, silica gel 60 (Kanto Chemicals) or Presep Silica Gel (WAKO) was used as the silica gel for purification used in silica gel column chromatography. Moreover, the silica gel for refinement | purification used for NH silica gel column chromatography used NH silica gel (Fuji Silisia Chemical LTD.) Or Hi-Flash Column Amino (YAMAZENE CORPORATION).

プロトン核磁気共鳴スペクトルの測定には、Varian Mercury 400、Varian Mercury Plus 400、Varian INOVA 500またはAvance600MHz(Bruker)を用い、特に記載のない限り、400メガヘルツで測定した。プロトン核磁気共鳴スペクトルの化学シフトは、テトラメチルシランに対するδ単位(ppm)で記録され、カップリング定数はヘルツ(Hz)で記録されている。分裂パターンの略号は以下の通りである。s:シングレット、d:ダブレット、t:トリプレット、m:マルチプレット、brs:ブロードシングレット。   The proton nuclear magnetic resonance spectrum was measured using a Varian Mercury 400, Varian Mercury Plus 400, Varian INOVA 500 or Avance 600 MHz (Bruker) at 400 MHz unless otherwise specified. The chemical shift of the proton nuclear magnetic resonance spectrum is recorded in δ units (ppm) relative to tetramethylsilane, and the coupling constant is recorded in hertz (Hz). The abbreviations of the division pattern are as follows. s: singlet, d: doublet, t: triplet, m: multiplet, brs: broad singlet.

製造例、実施例および参考例中、市販されている化合物は、適宜、市販品を用いた。   In the production examples, examples and reference examples, commercially available compounds were appropriately used as the commercially available compounds.

[製造例1−1]N−(4−クロロピリジン−2−イル)アセトアミド
市販の2−アミノ−4−クロロピリジン(50g、389mmol)を無水酢酸(500mL)に溶解し、20℃にてトリエチルアミン(271mL、1.94mol)を加え、60℃で12時間攪拌した。室温まで冷却した後に、溶媒を留去した。残渣をシリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=4:1〜1:1)で精製後、目的物画分を減圧濃縮し、標記化合物(66g、99%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 2.21 (3H, s), 7.05 (1H, dd, J=5.4, 1.9 Hz), 8.15 (1H, d, J=5.4 Hz), 8.30 (2H, brs).
[Production Example 1-1] N- (4-chloropyridin-2-yl) acetamide
Commercially available 2-amino-4-chloropyridine (50 g, 389 mmol) was dissolved in acetic anhydride (500 mL), triethylamine (271 mL, 1.94 mol) was added at 20 ° C., and the mixture was stirred at 60 ° C. for 12 hours. After cooling to room temperature, the solvent was distilled off. The residue was purified by silica gel column chromatography (n-heptane: ethyl acetate = 4: 1 to 1: 1), and the target fraction was concentrated under reduced pressure to obtain the title compound (66 g, 99%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 2.21 (3H, s), 7.05 (1H, dd, J = 5.4, 1.9 Hz), 8.15 (1H, d, J = 5.4 Hz), 8.30 (2H , brs).

[製造例1−2]フェニル メチルカーバメート
市販のメチルアミン塩酸塩(50g、0.74mol)、ピリジン(124mL、1.53mol)およびN,N−ジメチルホルムアミド(500mL)の混合物を5℃にて攪拌し、市販のクロロ炭酸フェニル(94mL、0.75mol)を2時間かけて滴下し加えた。滴下後、窒素雰囲気下、室温にて16時間攪拌した。反応混合物を氷水(2L)に加え、酢酸エチル(1.5L)で2回抽出を行った。有機層を水(1L)および飽和食塩水(300mL)で洗浄した。有機層を無水硫酸マグネシウムで乾燥後、溶媒を留去した。濃縮残渣にn−ヘプタンおよび酢酸エチルを加え、沈殿物をn−ヘプタンおよびtert−ブチルメチルエーテルを用いて濾取洗浄し、標記化合物(74.2g、66%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 2.90 (3H, d, J=4.9 Hz), 4.95 (1H, brs), 7.08-7.16 (2H, m), 7.16-7.24 (1H, m), 7.31-7.41 (2H, m).
[Production Example 1-2] Phenyl methyl carbamate
A mixture of commercially available methylamine hydrochloride (50 g, 0.74 mol), pyridine (124 mL, 1.53 mol) and N, N-dimethylformamide (500 mL) was stirred at 5 ° C., and commercially available phenyl chlorocarbonate (94 mL, 0.75 mol) was added dropwise over 2 hours. After dropping, the mixture was stirred at room temperature for 16 hours under a nitrogen atmosphere. The reaction mixture was added to ice water (2 L) and extracted twice with ethyl acetate (1.5 L). The organic layer was washed with water (1 L) and saturated brine (300 mL). The organic layer was dried over anhydrous magnesium sulfate and the solvent was distilled off. N-Heptane and ethyl acetate were added to the concentrated residue, and the precipitate was collected by filtration using n-heptane and tert-butyl methyl ether to obtain the title compound (74.2 g, 66%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 2.90 (3H, d, J = 4.9 Hz), 4.95 (1H, brs), 7.08-7.16 (2H, m), 7.16-7.24 (1H, m) , 7.31-7.41 (2H, m).

[製造例1−3]1−(4−フェニルピペリジン−1−イル)エタノン
市販の4−フェニルピペリジン(10g、62mmol)、ピリジン(5.7mL、70.5mmol)およびテトラヒドロフラン(80mL)の混合物を0℃にて攪拌し、塩化アセチル(5mL、70.3mmol)およびテトラヒドロフラン(20mL)の混合物を10分間かけて滴下した。その混合物を窒素雰囲気下、25℃にて14時間攪拌した。反応液に酢酸エチル(100mL)、水(100mL)を加え、分液した。水層を酢酸エチル(100mL)で抽出した後、有機層を合わせ、飽和炭酸水素ナトリウム水溶液(100mL)、水(100mL)、続いて飽和食塩水(50mL)で洗浄した。有機層を無水硫酸マグネシウムで乾燥後、溶媒を留去し、標記化合物(12.3g、98%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 1.52-1.78 (2H, m), 1.81-1.99 (2H, m), 2.14 (3H, s), 2.63 (1H, td, J=12.9, 2.7 Hz), 2.74 (1H, tt, J=12.1, 3.7 Hz), 3.17 (1H, td, J=13.2, 2.6 Hz), 3.84-4.02 (1H, m), 4.69-4.89 (1H, m), 7.08-7.43 (5H, m).
[Production Example 1-3] 1- (4-phenylpiperidin-1-yl) ethanone
A mixture of commercially available 4-phenylpiperidine (10 g, 62 mmol), pyridine (5.7 mL, 70.5 mmol) and tetrahydrofuran (80 mL) was stirred at 0 ° C., and acetyl chloride (5 mL, 70.3 mmol) and tetrahydrofuran (20 mL) were stirred. ) Was added dropwise over 10 minutes. The mixture was stirred at 25 ° C. for 14 hours under a nitrogen atmosphere. Ethyl acetate (100 mL) and water (100 mL) were added to the reaction solution and the phases were separated. The aqueous layer was extracted with ethyl acetate (100 mL), and the organic layers were combined and washed with saturated aqueous sodium hydrogen carbonate solution (100 mL), water (100 mL), and then saturated brine (50 mL). The organic layer was dried over anhydrous magnesium sulfate and the solvent was distilled off to obtain the title compound (12.3 g, 98%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 1.52-1.78 (2H, m), 1.81-1.99 (2H, m), 2.14 (3H, s), 2.63 (1H, td, J = 12.9, 2.7 Hz), 2.74 (1H, tt, J = 12.1, 3.7 Hz), 3.17 (1H, td, J = 13.2, 2.6 Hz), 3.84-4.02 (1H, m), 4.69-4.89 (1H, m), 7.08 -7.43 (5H, m).

[製造例1−4]4−(1−アセチルピペリジン−4−イル)ベンゾイック アシッド
塩化アルミニウム(III)(26g、195mmol)およびジクロロメタン(200mL)の混合物を0℃にて攪拌し、塩化オキサリル(20mL、228mmol)を10分間かけ滴下した。続いて、製造例1−3に記載の1−(4−フェニルピペリジン−1−イル)エタノン(12.3g、60.5mmol)およびジクロロメタン(50mL)の混合物を30分間かけ滴下した。その混合物を窒素雰囲気下、25℃にて14時間攪拌した。反応液を氷にあけ、酢酸エチル(1L)、水(1L)を加え、分液した。水層を2回酢酸エチル(1L)で抽出した後、有機層を水(1L)で二回、続いて飽和食塩水(500mL)で洗浄した。有機層を無水硫酸マグネシウムで乾燥後、溶媒を留去した。濃縮残渣に酢酸エチルを加え、生成物を酢酸エチルを用いて濾取洗浄し、標記化合物(9.09g、61%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 1.49-1.82 (2H, m), 1.92 (2H, t, J=13.2 Hz), 2. 15 (3H, s), 2.65 (1H, t, J=11.7 Hz), 2.75-2.94 (1H, m), 3.08-3.30 (1H, m), 3.97 (1H, d, J=13.2 Hz), 4.82 (1H, d, J=12.8 Hz), 7.30 (2H, d, J=8.4 Hz), 8.05 (2H, d, J=8.1 Hz).
[Production Example 1-4] 4- (1-acetylpiperidin-4-yl) benzoic acid
A mixture of aluminum (III) chloride (26 g, 195 mmol) and dichloromethane (200 mL) was stirred at 0 ° C., and oxalyl chloride (20 mL, 228 mmol) was added dropwise over 10 minutes. Subsequently, a mixture of 1- (4-phenylpiperidin-1-yl) ethanone (12.3 g, 60.5 mmol) and dichloromethane (50 mL) described in Production Example 1-3 was added dropwise over 30 minutes. The mixture was stirred at 25 ° C. for 14 hours under a nitrogen atmosphere. The reaction mixture was poured into ice, and ethyl acetate (1 L) and water (1 L) were added to separate the layers. The aqueous layer was extracted twice with ethyl acetate (1 L), and then the organic layer was washed twice with water (1 L) followed by saturated brine (500 mL). The organic layer was dried over anhydrous magnesium sulfate and the solvent was distilled off. Ethyl acetate was added to the concentrated residue, and the product was filtered and washed with ethyl acetate to obtain the title compound (9.09 g, 61%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 1.49-1.82 (2H, m), 1.92 (2H, t, J = 13.2 Hz), 2. 15 (3H, s), 2.65 (1H, t, J = 11.7 Hz), 2.75-2.94 (1H, m), 3.08-3.30 (1H, m), 3.97 (1H, d, J = 13.2 Hz), 4.82 (1H, d, J = 12.8 Hz), 7.30 ( 2H, d, J = 8.4 Hz), 8.05 (2H, d, J = 8.1 Hz).

[製造例1−5]4−(ピペリジン−4−イル)ベンゾイック アシッド塩酸塩
製造例1−4に記載の4−(1−アセチルピペリジン−4−イル)ベンゾイック アシッド(4.50g、18.2mmol)および5M塩酸(50mL、250mmol)の混合物を窒素雰囲気下、140℃にて18時間攪拌した。室温に冷却後、生成物を濾取、水で洗浄し、標記化合物(3.77g、86%)を得た。
1H-NMR Spectrum (DMSO-d6) δ(ppm): 1.60-2.15 (4H, m), 2.76-3.16 (3H, m), 3.27-3.45 (2H, m), 7.36 (2H, d, J=8.1 Hz), 7.92 (2H, d, J=8.1 Hz), 8.65-9.04 (2H, m), 12.89 (1H, brs).
[Production Example 1-5] 4- (Piperidin-4-yl) benzoic acid hydrochloride
A mixture of 4- (1-acetylpiperidin-4-yl) benzoic acid (4.50 g, 18.2 mmol) and 5M hydrochloric acid (50 mL, 250 mmol) described in Production Example 1-4 at 140 ° C. under a nitrogen atmosphere. Stir for 18 hours. After cooling to room temperature, the product was collected by filtration and washed with water to give the title compound (3.77 g, 86%).
1 H-NMR Spectrum (DMSO-d 6 ) δ (ppm): 1.60-2.15 (4H, m), 2.76-3.16 (3H, m), 3.27-3.45 (2H, m), 7.36 (2H, d, J = 8.1 Hz), 7.92 (2H, d, J = 8.1 Hz), 8.65-9.04 (2H, m), 12.89 (1H, brs).

[製造例1−6]4−(1−(tert−ブトキシカルボニル)ピペリジン−4−イル)ベンゾイック アシッド
製造例1−5に記載の4−(ピペリジン−4−イル)ベンゾイック アシッド塩酸塩(2.00g、8.27mmol)、1M水酸化ナトリウム溶液(25mL、25mmol)およびアセトン(50mL)の混合物を25℃にて攪拌し、ジ−tert−ブチル ジカーボネート(1.9g、8.71mmol)のアセトン(25mL)溶液を10分間かけ滴下し加えた。混合物を窒素雰囲気下、25℃にて18時間攪拌した。0℃にて冷却下、1M塩酸(17mL)を加えた。酢酸エチル(100mL)で2回抽出を行った。有機層を飽和食塩水(50mL)で洗浄した。有機層を無水硫酸マグネシウムで乾燥後、減圧濃縮した。濃縮残渣にn−ヘプタンおよびtert−ブチルメチルエーテルを加え、生成物をn−ヘプタンを用いて濾取洗浄し、標記化合物(2.30g、91%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 1.49 (9H, s), 1.57-1.76 (2H, m), 1.84 (2H, d, J=13.5 Hz), 2.62-2.97 (3H, m), 4.27 (2H, brs), 7.28-7.36 (2H, m), 7.98-8.10 (2H, m).
[Production Example 1-6] 4- (1- (tert-butoxycarbonyl) piperidin-4-yl) benzoic acid
A mixture of 4- (piperidin-4-yl) benzoic acid hydrochloride (2.00 g, 8.27 mmol), 1M sodium hydroxide solution (25 mL, 25 mmol) and acetone (50 mL) described in Preparation Example 1-5 was added. The mixture was stirred at 0 ° C., and a solution of di-tert-butyl dicarbonate (1.9 g, 8.71 mmol) in acetone (25 mL) was added dropwise over 10 minutes. The mixture was stirred at 25 ° C. for 18 hours under a nitrogen atmosphere. Under cooling at 0 ° C., 1M hydrochloric acid (17 mL) was added. Extraction was performed twice with ethyl acetate (100 mL). The organic layer was washed with saturated brine (50 mL). The organic layer was dried over anhydrous magnesium sulfate and concentrated under reduced pressure. N-Heptane and tert-butyl methyl ether were added to the concentrated residue, and the product was filtered and washed with n-heptane to obtain the title compound (2.30 g, 91%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 1.49 (9H, s), 1.57-1.76 (2H, m), 1.84 (2H, d, J = 13.5 Hz), 2.62-2.97 (3H, m) , 4.27 (2H, brs), 7.28-7.36 (2H, m), 7.98-8.10 (2H, m).

[製造例1−7]3−ヒドロキシ−4−(2−メトキシエトキシ)ベンズアルデヒド
市販の3,4−ジヒドロキシベンズアルデヒド(39.3g、285mmol)と炭酸ナトリウム(45.2g、427mmol)をN,N−ジメチルホルムアミド(400mL)に溶解させた後、窒素雰囲気下、室温で市販の2−ブロモエチル メチル エーテル(26.7mL、285mmol)を加え、5日間攪拌した。0℃に冷却後、2M塩酸、酢酸エチル、水を加え分配した。水層を酢酸エチルで抽出し、合わせた有機層を飽和食塩水で洗浄し、無水硫酸マグネシウムで乾燥後、濾過した。溶媒を留去し、ジクロロメタンを加えた後、析出物を濾別し、得られた濾液をシリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=17:3〜1:1)で精製した。目的物画分を減圧濃縮し、標記化合物(12.9g、23%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 3.47 (3H, s), 3.76-3.80 (2H, m), 4.25-4.29 (2H, m), 6.40 (1H, brs), 7.01 (1H, d, J=8.4 Hz), 7.41 (1H, dd, J=8.2, 2.0 Hz), 7.45 (1H, d, J=1.8 Hz), 9.85 (1H, s).
[Production Example 1-7] 3-Hydroxy-4- (2-methoxyethoxy) benzaldehyde
Commercially available 3,4-dihydroxybenzaldehyde (39.3 g, 285 mmol) and sodium carbonate (45.2 g, 427 mmol) were dissolved in N, N-dimethylformamide (400 mL), and then commercially available at room temperature under a nitrogen atmosphere. -Bromoethyl methyl ether (26.7 mL, 285 mmol) was added and stirred for 5 days. After cooling to 0 ° C., 2M hydrochloric acid, ethyl acetate and water were added and partitioned. The aqueous layer was extracted with ethyl acetate, and the combined organic layer was washed with saturated brine, dried over anhydrous magnesium sulfate, and then filtered. After the solvent was distilled off and dichloromethane was added, the precipitate was separated by filtration, and the obtained filtrate was purified by silica gel column chromatography (n-heptane: ethyl acetate = 17: 3 to 1: 1). The target fraction was concentrated under reduced pressure to obtain the title compound (12.9 g, 23%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 3.47 (3H, s), 3.76-3.80 (2H, m), 4.25-4.29 (2H, m), 6.40 (1H, brs), 7.01 (1H, d, J = 8.4 Hz), 7.41 (1H, dd, J = 8.2, 2.0 Hz), 7.45 (1H, d, J = 1.8 Hz), 9.85 (1H, s).

[製造例1−8]3−(ベンジルオキシ)−4−(2−メトキシエトキシ)ベンズアルデヒド
製造例1−7に記載の3−ヒドロキシ−4−(2−メトキシエトキシ)ベンズアルデヒド(12.9g、65.9mmol)およびエタノール(130mL)の混合物に、窒素雰囲気下、室温で炭酸カリウム(11.8g、85.7mmol)、塩化ベンジル(10mL、86.9mmol)を加え、90℃にて2時間加熱攪拌した。0℃に冷却後、2M塩酸、酢酸エチル、水を加え分配した。有機層を飽和食塩水で洗浄し、無水硫酸マグネシウムで乾燥後、濾過した。溶媒を留去し、得られた残渣をシリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=9:1〜1:1)で精製した。目的物画分を減圧濃縮し、標記化合物(17.6g、93%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 3.46 (3H, s), 3.79-3.85 (2H, m), 4.24-4.30 (2H, m), 5.18 (2H, s), 7.03 (1H, d, J=8.1 Hz), 7.29-7.35 (1H, m), 7.35-7.41 (2H, m), 7.43-7.50 (4H, m), 9.82 (1H, s).
[Production Example 1-8] 3- (Benzyloxy) -4- (2-methoxyethoxy) benzaldehyde
To a mixture of 3-hydroxy-4- (2-methoxyethoxy) benzaldehyde (12.9 g, 65.9 mmol) and ethanol (130 mL) described in Production Example 1-7, potassium carbonate (11. 8 g, 85.7 mmol) and benzyl chloride (10 mL, 86.9 mmol) were added, and the mixture was heated and stirred at 90 ° C. for 2 hours. After cooling to 0 ° C., 2M hydrochloric acid, ethyl acetate and water were added and partitioned. The organic layer was washed with saturated brine, dried over anhydrous magnesium sulfate, and filtered. The solvent was distilled off, and the resulting residue was purified by silica gel column chromatography (n-heptane: ethyl acetate = 9: 1 to 1: 1). The target fraction was concentrated under reduced pressure to obtain the title compound (17.6 g, 93%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 3.46 (3H, s), 3.79-3.85 (2H, m), 4.24-4.30 (2H, m), 5.18 (2H, s), 7.03 (1H, d, J = 8.1 Hz), 7.29-7.35 (1H, m), 7.35-7.41 (2H, m), 7.43-7.50 (4H, m), 9.82 (1H, s).

[製造例1−9](E)−2−(ベンジルオキシ)−1−(2−メトキシエトキシ)−4−(2−ニトロビニル)ベンゼン
製造例1−8に記載の3−(ベンジルオキシ)−4−(2−メトキシエトキシ)ベンズアルデヒド(17.6g、61.5mmol)を酢酸(49.3mL)に溶解させた後、窒素雰囲気下、室温で酢酸アンモニウム(5.69g、73.8mmol)、ニトロメタン(8.32mL、154mmol)を加え、130℃にて2時間加熱攪拌した。室温まで冷却後、析出物を濾取、エタノールにて洗浄し、標記化合物を定量的に得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 3.46 (3H, s), 3.78-3.84 (2H, m), 4.21-4.27 (2H, m), 5.16 (2H, s), 6.97 (1H, d, J=8.4 Hz), 7.06 (1H, d, J=1.8 Hz), 7.16 (1H, dd, J=8.4, 2.2 Hz), 7.30-7.48 (6H, m), 7.91 (1H, d, J=13.5 Hz).
[Production Example 1-9] (E) -2- (benzyloxy) -1- (2-methoxyethoxy) -4- (2-nitrovinyl) benzene
3- (Benzyloxy) -4- (2-methoxyethoxy) benzaldehyde (17.6 g, 61.5 mmol) described in Production Example 1-8 was dissolved in acetic acid (49.3 mL). Ammonium acetate (5.69 g, 73.8 mmol) and nitromethane (8.32 mL, 154 mmol) were added at room temperature, and the mixture was heated with stirring at 130 ° C. for 2 hours. After cooling to room temperature, the precipitate was collected by filtration and washed with ethanol to give the title compound quantitatively.
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 3.46 (3H, s), 3.78-3.84 (2H, m), 4.21-4.27 (2H, m), 5.16 (2H, s), 6.97 (1H, d, J = 8.4 Hz), 7.06 (1H, d, J = 1.8 Hz), 7.16 (1H, dd, J = 8.4, 2.2 Hz), 7.30-7.48 (6H, m), 7.91 (1H, d, J = 13.5 Hz).

[製造例1−10]6−(2−メトキシエトキシ)−1H−インドール−5−オール
製造例1−9に記載の(E)−2−(ベンジルオキシ)−1−(2−メトキシエトキシ)−4−(2−ニトロビニル)ベンゼン(20.2g、61.5mmol)および酢酸(120mL)の混合物に、25℃にて69%硝酸(15mL、233mmol)を加え、室温で6時間攪拌した。反応混合物を氷にあけ、沈殿物を濾取後、水で洗浄し、粗生成物(23.0g)を得た。
粗生成物(23.0g)をメタノール(500mL)に懸濁させた後、室温で10%パラジウム−炭素(50%含水品)(8g)を加えて水素雰囲気下で6時間攪拌した。触媒をセライトを用いて濾過後、濾液を減圧濃縮し、シリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=2:1〜1:1)で精製した。目的物画分を減圧濃縮し、標記化合物(3.94g、31%)を得た。
1H-NMR Spectrum (CDCl3) δ(ppm): 3.48 (3H, s), 3.69-3.78 (2H, m), 4.16-4.23 (2H, m), 6.24 (1H, s), 6.41 (1H, ddd, J=3.1, 2.1, 0.8 Hz), 6.97 (1H, s), 7.10 (1H, dd, J=3.2, 2.5 Hz), 7.15 (1H, s), 7.94 (1H, brs).
[Production Example 1-10] 6- (2-methoxyethoxy) -1H-indole-5-ol
(E) -2- (benzyloxy) -1- (2-methoxyethoxy) -4- (2-nitrovinyl) benzene (20.2 g, 61.5 mmol) and acetic acid (120 mL) described in Preparation Example 1-9 69% nitric acid (15 mL, 233 mmol) was added to the mixture at 25 ° C., and the mixture was stirred at room temperature for 6 hours. The reaction mixture was poured into ice, and the precipitate was collected by filtration and washed with water to obtain a crude product (23.0 g).
The crude product (23.0 g) was suspended in methanol (500 mL), 10% palladium-carbon (50% water-containing product) (8 g) was added at room temperature, and the mixture was stirred under a hydrogen atmosphere for 6 hr. The catalyst was filtered through celite, and the filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (n-heptane: ethyl acetate = 2: 1 to 1: 1). The target fraction was concentrated under reduced pressure to obtain the title compound (3.94 g, 31%).
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 3.48 (3H, s), 3.69-3.78 (2H, m), 4.16-4.23 (2H, m), 6.24 (1H, s), 6.41 (1H, ddd, J = 3.1, 2.1, 0.8 Hz), 6.97 (1H, s), 7.10 (1H, dd, J = 3.2, 2.5 Hz), 7.15 (1H, s), 7.94 (1H, brs).

[製造例1−11]N−(4−((6−(2−メトキシエトキシ)−1H−インドール−5−イル)オキシ)ピリジン−2−イル)アセトアミド
製造例1−10に記載の6−(2−メトキシエトキシ)−1H−インドール−5−オール(3.94g、19.0mmol)、製造例1−1に記載のN−(4−クロロピリジン−2−イル)アセトアミド(3.25g、19.0mmol)をジメチルスルホキシド(25mL)に溶解させ、室温にて97%カリウム tert−ブトキシド(2.20g、19.0mmol)を加え、150℃にて13時間加熱攪拌した。反応液に室温で水と酢酸エチルを加え分配した。水層を酢酸エチルで3回抽出し、合わせた有機層を水で洗浄した。有機層を無水硫酸ナトリウムで乾燥した。乾燥剤を濾過し、濾液を減圧濃縮後、NHシリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=2:3〜0:1〜酢酸エチル:メタノール=49:1〜9:1)で精製した。目的物画分を減圧濃縮し、標記化合物(3.45g、53%)を得た。
1H-NMR Spectrum (500MHz, CDCl3) δ(ppm): 2.13 (3H, s), 3.27 (3H, s), 3.54-3.58 (2H, m), 4.07-4.11 (2H, m), 6.46-6.50 (1H, m), 6.54 (1H, dd, J=5.8, 1.9 Hz), 7.05 (1H, s), 7.14-7.17 (1H, m), 7.36 (1H, s), 7.75 (1H, brs), 8.02 (1H, d, J=5.8 Hz), 8.10 (1H, brs), 8.19 (1H, brs).
[Production Example 1-11] N- (4-((6- (2-methoxyethoxy) -1H-indol-5-yl) oxy) pyridin-2-yl) acetamide
6- (2-methoxyethoxy) -1H-indole-5-ol (3.94 g, 19.0 mmol) described in Preparation Example 1-10, N- (4-chloropyridine- described in Preparation Example 1-1 2-yl) acetamide (3.25 g, 19.0 mmol) was dissolved in dimethyl sulfoxide (25 mL), 97% potassium tert-butoxide (2.20 g, 19.0 mmol) was added at room temperature, and 13 at 150 ° C. Stir with heating for hours. The reaction solution was partitioned by adding water and ethyl acetate at room temperature. The aqueous layer was extracted 3 times with ethyl acetate, and the combined organic layers were washed with water. The organic layer was dried over anhydrous sodium sulfate. The desiccant was filtered, and the filtrate was concentrated under reduced pressure, and then purified by NH silica gel column chromatography (n-heptane: ethyl acetate = 2: 3 to 0: 1 to ethyl acetate: methanol = 49: 1 to 9: 1). The target fraction was concentrated under reduced pressure to obtain the title compound (3.45 g, 53%).
1 H-NMR Spectrum (500MHz, CDCl 3 ) δ (ppm): 2.13 (3H, s), 3.27 (3H, s), 3.54-3.58 (2H, m), 4.07-4.11 (2H, m), 6.46- 6.50 (1H, m), 6.54 (1H, dd, J = 5.8, 1.9 Hz), 7.05 (1H, s), 7.14-7.17 (1H, m), 7.36 (1H, s), 7.75 (1H, brs) , 8.02 (1H, d, J = 5.8 Hz), 8.10 (1H, brs), 8.19 (1H, brs).

[製造例1−12]4−((6−(2−メトキシエトキシ)−1H−インドール−5−イル)オキシ)ピリジン−2−アミン
製造例1−11に記載のN−(4−((6−(2−メトキシエトキシ)−1H−インドール−5−イル)オキシ)ピリジン−2−イル)アセトアミド(3.45g、10.1mmol)をメタノール(50mL)に溶解させ、室温にて2M水酸化ナトリウム溶液(50mL)を加えて70℃にて3時間加熱攪拌した。反応混合物に、水と酢酸エチルを加え分配した。水層を酢酸エチルで3回抽出し、合わせた有機層を無水硫酸ナトリウムで乾燥した。乾燥剤を濾過し、濾液を減圧濃縮し、NHシリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=3:7〜0:1〜酢酸エチル:メタノール=49:1〜24:1)で精製した。目的物画分、混合物画分を別々に減圧濃縮し、混合物画分をシリカゲルカラムクロマトグラフィー(酢酸エチル:メタノール=1:0〜9:1)で再度精製し、先の目的物画分と合わせて標記化合物(2.60g、86%)を得た。
1H-NMR Spectrum (500MHz, CDCl3) δ(ppm): 3.31 (3H, s), 3.58-3.63 (2H, m), 4.08-4.11 (2H, m), 4.28 (2H, brs), 5.90 (1H, d, J=2.4 Hz), 6.29 (1H, dd, J=6.1, 2.2 Hz), 6.44-6.52 (1H, m), 7.06 (1H, s), 7.15-7.20 (1H, m), 7.34 (1H, s), 7.88 (1H, d, J=5.8 Hz), 8.22 (1H, brs).
[Production Example 1-12] 4-((6- (2-methoxyethoxy) -1H-indol-5-yl) oxy) pyridin-2-amine
N- (4-((6- (2-methoxyethoxy) -1H-indol-5-yl) oxy) pyridin-2-yl) acetamide (3.45 g, 10.1 mmol) described in Preparation Example 1-11 Was dissolved in methanol (50 mL), 2M sodium hydroxide solution (50 mL) was added at room temperature, and the mixture was stirred with heating at 70 ° C. for 3 hr. The reaction mixture was partitioned by adding water and ethyl acetate. The aqueous layer was extracted 3 times with ethyl acetate, and the combined organic layers were dried over anhydrous sodium sulfate. The desiccant was filtered, the filtrate was concentrated under reduced pressure, and purified by NH silica gel column chromatography (n-heptane: ethyl acetate = 3: 7 to 0: 1 to ethyl acetate: methanol = 49: 1 to 24: 1). The target fraction and the mixture fraction were separately concentrated under reduced pressure, and the mixture fraction was purified again by silica gel column chromatography (ethyl acetate: methanol = 1: 0 to 9: 1) and combined with the previous target fraction. This gave the title compound (2.60 g, 86%).
1 H-NMR Spectrum (500MHz, CDCl 3 ) δ (ppm): 3.31 (3H, s), 3.58-3.63 (2H, m), 4.08-4.11 (2H, m), 4.28 (2H, brs), 5.90 ( 1H, d, J = 2.4 Hz), 6.29 (1H, dd, J = 6.1, 2.2 Hz), 6.44-6.52 (1H, m), 7.06 (1H, s), 7.15-7.20 (1H, m), 7.34 (1H, s), 7.88 (1H, d, J = 5.8 Hz), 8.22 (1H, brs).

[製造例1−13]5−[(2−アミノピリジン−4−イル)オキシ]−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド
製造例1−12に記載の4−((6−(2−メトキシエトキシ)−1H−インドール−5−イル)オキシ)ピリジン−2−アミン(2.60g、8.67mmol)をN,N−ジメチルホルムアミド(50mL)に溶解し、窒素雰囲気下、室温にて50−72%水素化ナトリウム 油状(499mg)を加えた。製造例1−2に記載のフェニル メチルカーバメート(1.97g、13.0mmol)を加え室温で1時間攪拌した。反応混合物を0℃に冷却し、酢酸エチル、水を加え分配した。水層を酢酸エチルで2回抽出し、水層に塩化ナトリウムを加えて酢酸エチルで3回抽出した。合わせた有機層を無水硫酸ナトリウムで乾燥した。乾燥剤を濾過し、濾液を減圧濃縮し、NHシリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=1:4〜0:1〜酢酸エチル:メタノール=49:1〜24:1)で精製した。目的物画分を減圧濃縮後、酢酸エチルを加えて析出物を濾取、洗浄し、標記化合物(2.23g、72%)を得た。
1H-NMR Spectrum (500MHz, CDCl3) δ(ppm): 3.06 (3H, d, J=4.9 Hz), 3.29 (3H, s), 3.59-3.63 (2H, m), 4.14-4.17 (2H, m), 4.30 (2H, brs), 5.52-5.59 (1H, m), 5.89 (1H, d, J=2.4 Hz), 6.27 (1H, dd, J=5.8, 1.9 Hz), 6.55 (1H, d, J=3.9 Hz), 7.27-7.29 (2H, m), 7.89 (1H, d, J=5.9 Hz), 7.99 (1H, s).
[Production Example 1-13] 5-[(2-Aminopyridin-4-yl) oxy] -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide
4-((6- (2-methoxyethoxy) -1H-indol-5-yl) oxy) pyridin-2-amine (2.60 g, 8.67 mmol) described in Preparation Example 1-12 was converted to N, N- Dissolved in dimethylformamide (50 mL), 50-72% sodium hydride oil (499 mg) was added at room temperature under nitrogen atmosphere. Phenyl methyl carbamate (1.97 g, 13.0 mmol) described in Production Example 1-2 was added, and the mixture was stirred at room temperature for 1 hour. The reaction mixture was cooled to 0 ° C., and partitioned by adding ethyl acetate and water. The aqueous layer was extracted twice with ethyl acetate, sodium chloride was added to the aqueous layer and extracted three times with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate. The desiccant was filtered, the filtrate was concentrated under reduced pressure, and purified by NH silica gel column chromatography (n-heptane: ethyl acetate = 1: 4 to 0: 1 to ethyl acetate: methanol = 49: 1 to 24: 1). The target fraction was concentrated under reduced pressure, ethyl acetate was added, and the precipitate was collected by filtration and washed to obtain the title compound (2.23 g, 72%).
1 H-NMR Spectrum (500MHz, CDCl 3 ) δ (ppm): 3.06 (3H, d, J = 4.9 Hz), 3.29 (3H, s), 3.59-3.63 (2H, m), 4.14-4.17 (2H, m), 4.30 (2H, brs), 5.52-5.59 (1H, m), 5.89 (1H, d, J = 2.4 Hz), 6.27 (1H, dd, J = 5.8, 1.9 Hz), 6.55 (1H, d , J = 3.9 Hz), 7.27-7.29 (2H, m), 7.89 (1H, d, J = 5.9 Hz), 7.99 (1H, s).

[製造例1−14]6−(2−メトキシエトキシ)−N−メチル−5−{[2−({[4−(ピペリジン−4−イル)フェニル]カルボニル}アミノ)ピリジン−4−イル]オキシ}−1H−インドール−1−カルボキサミド
ベンゾトリアゾール(609mg、5.11mmol)をジクロロメタン(25mL)に溶解させ、窒素雰囲気下、室温にて塩化チオニル(373μL、5.11mmol)を加え5分間攪拌した。反応混合物に製造例1−6に記載の4−(1−(tert−ブトキシカルボニル)ピペリジン−4−イル)ベンゾイック アシッド(1.3g、4.26mmol)を室温にて加え、30分間攪拌した。反応混合物を無水硫酸ナトリウムを敷き詰めたグラスフィルターで濾過し、ジクロロメタンで洗浄し、濾液を製造例1−13に記載の5−[(2−アミノピリジン−4−イル)オキシ]−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド(0.95g、2.67mmol)、トリエチルアミン(1.86mL、13.3mmol)、4−ジメチルアミノピリジン(16mg、0.133mmol)、N,N−ジメチルホルムアミド(3mL)およびジクロロメタン(20mL)の混合物に0℃にて5分かけて加え、ジクロロメタン(10mL)で洗い込み同温で5分間攪拌した。室温で2時間攪拌後、40%メチルアミン水溶液(2.3mL、26.7mmol)を加え室温で1.5時間攪拌した。反応混合物に飽和炭酸水素ナトリウム水溶液を加え分配し、水層を酢酸エチルで3回抽出した。合わせた有機層を無水硫酸ナトリウムで乾燥した。乾燥剤を濾過後、濾液を減圧濃縮し、シリカゲルカラムクロマトグラフィー(n−ヘプタン:酢酸エチル=1:1〜0:1〜酢酸エチル:メタノール=49:1〜23:2)で精製し、粗生成物(1.11g)を得た。
粗生成物(1.11g)をジクロロメタン(50mL)に溶解させ、室温でトリフルオロ酢酸(5.0mL)を加えた。室温にて30分間攪拌後、減圧濃縮し、残渣をジクロロメタンとトリエチルアミンに溶解させ、減圧濃縮した。残渣をNHシリカゲルカラムクロマトグラフィー(酢酸エチル:メタノール=1:0〜22:3)で精製し、標記化合物(829mg、57%)を得た。
1H-NMR Spectrum (500MHz, CDCl3) δ(ppm): 1.59-1.69 (2H, m), 1.83 (2H, d, J=14.1 Hz), 2.68 (1H, tt, J=12.0, 3.6 Hz), 2.75 (2H, td, J=12.2, 2.4 Hz), 3.04 (3H, d, J=4.9 Hz), 3.17-3.23 (2H, m), 3.26 (3H, s), 3.55-3.61 (2H, m), 4.15-4.21 (2H, m), 5.57-5.65 (1H, m), 6.53 (1H, d, J=3.4 Hz), 6.62 (1H, dd, J=5.8, 2.4 Hz), 7.25 (1H, d, J=3.9 Hz), 7.30-7.34 (3H, m), 7.77-7.82 (2H, m), 7.91 (1H, d, J=2.4 Hz), 8.02 (1H, s), 8.10 (1H, d, J=5.9 Hz), 8.50 (1H, brs).
[Production Example 1-14] 6- (2-methoxyethoxy) -N-methyl-5-{[2-({[4- (piperidin-4-yl) phenyl] carbonyl} amino) pyridin-4-yl] Oxy} -1H-indole-1-carboxamide
Benzotriazole (609 mg, 5.11 mmol) was dissolved in dichloromethane (25 mL), thionyl chloride (373 μL, 5.11 mmol) was added at room temperature under a nitrogen atmosphere, and the mixture was stirred for 5 min. 4- (1- (tert-Butoxycarbonyl) piperidin-4-yl) benzoic acid (1.3 g, 4.26 mmol) described in Production Example 1-6 was added to the reaction mixture at room temperature, and the mixture was stirred for 30 minutes. The reaction mixture was filtered through a glass filter lined with anhydrous sodium sulfate, washed with dichloromethane, and the filtrate was prepared according to 5-[(2-aminopyridin-4-yl) oxy] -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide (0.95 g, 2.67 mmol), triethylamine (1.86 mL, 13.3 mmol), 4-dimethylaminopyridine (16 mg, 0.133 mmol), The mixture was added to a mixture of N, N-dimethylformamide (3 mL) and dichloromethane (20 mL) at 0 ° C. over 5 minutes, washed with dichloromethane (10 mL), and stirred at the same temperature for 5 minutes. After stirring at room temperature for 2 hours, 40% aqueous methylamine solution (2.3 mL, 26.7 mmol) was added, and the mixture was stirred at room temperature for 1.5 hours. A saturated aqueous sodium hydrogen carbonate solution was added to the reaction mixture for partitioning, and the aqueous layer was extracted three times with ethyl acetate. The combined organic layers were dried over anhydrous sodium sulfate. After filtering the desiccant, the filtrate was concentrated under reduced pressure and purified by silica gel column chromatography (n-heptane: ethyl acetate = 1: 1 to 0: 1 to ethyl acetate: methanol = 49: 1 to 23: 2) and crude. The product (1.11 g) was obtained.
The crude product (1.11 g) was dissolved in dichloromethane (50 mL) and trifluoroacetic acid (5.0 mL) was added at room temperature. The mixture was stirred at room temperature for 30 minutes and concentrated under reduced pressure. The residue was dissolved in dichloromethane and triethylamine and concentrated under reduced pressure. The residue was purified by NH silica gel column chromatography (ethyl acetate: methanol = 1: 0 to 22: 3) to obtain the title compound (829 mg, 57%).
1 H-NMR Spectrum (500MHz, CDCl 3 ) δ (ppm): 1.59-1.69 (2H, m), 1.83 (2H, d, J = 14.1 Hz), 2.68 (1H, tt, J = 12.0, 3.6 Hz) , 2.75 (2H, td, J = 12.2, 2.4 Hz), 3.04 (3H, d, J = 4.9 Hz), 3.17-3.23 (2H, m), 3.26 (3H, s), 3.55-3.61 (2H, m ), 4.15-4.21 (2H, m), 5.57-5.65 (1H, m), 6.53 (1H, d, J = 3.4 Hz), 6.62 (1H, dd, J = 5.8, 2.4 Hz), 7.25 (1H, d, J = 3.9 Hz), 7.30-7.34 (3H, m), 7.77-7.82 (2H, m), 7.91 (1H, d, J = 2.4 Hz), 8.02 (1H, s), 8.10 (1H, d , J = 5.9 Hz), 8.50 (1H, brs).

[製造例1−15]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド
製造例1−14に記載の6−(2−メトキシエトキシ)−N−メチル−5−{[2−({[4−(ピペリジン−4−イル)フェニル]カルボニル}アミノ)ピリジン−4−イル]オキシ}−1H−インドール−1−カルボキサミド(100mg、0.18mmol)およびテトラヒドロフラン(4mL)の混合物に、室温にてトリアセトキシ水素化ほう素ナトリウム(114mg、0.54mmol)と市販の2−ヒドロキシアセトアルデヒド(34.4mg、0.57mmol)を加え、室温にて2時間攪拌した。反応混合物に飽和炭酸水素ナトリウム水溶液、酢酸エチルを加え分配した。水層を酢酸エチルで抽出し、合わせた有機層を飽和食塩水で洗浄し、無水硫酸ナトリウムで乾燥後、濾過した。溶媒を留去し、得られた残渣をNHシリカゲルカラムクロマトグラフィー(酢酸エチル:メタノール=1:0〜97:3〜9:1)で精製した。目的物画分を減圧濃縮後、析出物をジエチルエーテルとn−ヘキサンの混合液で濾取、洗浄し、標記化合物(90mg、83%)を得た。得られた化合物の代表的な粉末X線回折角度を示す(化合物(I)・フリー体の結晶(Free Form A))。(2θ±0.2°):10.4°、10.9°、11.4°、13.5°、16.1°、19.7°、20.4°、21.5°、23.3°、および24.3°。
1H-NMR Spectrum (CDCl3) δ(ppm): 1.70-1.92 (4H, m), 2.15-2.24 (2H, m), 2.53-2.65 (3H, m), 3.01-3.09 (5H, m), 3.26 (3H, s), 3.56-3.60 (2H, m), 3.64 (2H, t, J=5.2 Hz), 4.15-4.20 (2H, m), 5.49-5.54 (1H, m), 6.55 (1H, d, J=3.7 Hz), 6.61 (1H, dd, J=5.8, 2.3 Hz), 7.24-7.28 (1H, m), 7.30-7.35 (3H, m), 7.81 (2H, d, J=8.2 Hz), 7.91 (1H, d, J=2.4 Hz), 8.01 (1H, s), 8.10 (1H, d, J=5.9 Hz), 8.50 (1H, brs).
[Production Example 1-15] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 (2-Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide
6- (2-methoxyethoxy) -N-methyl-5-{[2-({[4- (piperidin-4-yl) phenyl] carbonyl} amino) pyridin-4-yl described in Preparation Example 1-14 ] Oxy} -1H-indole-1-carboxamide (100 mg, 0.18 mmol) and tetrahydrofuran (4 mL) were mixed with sodium triacetoxyborohydride (114 mg, 0.54 mmol) and commercially available 2-hydroxy at room temperature. Acetaldehyde (34.4 mg, 0.57 mmol) was added and stirred at room temperature for 2 hours. The reaction mixture was partitioned by adding a saturated aqueous sodium hydrogen carbonate solution and ethyl acetate. The aqueous layer was extracted with ethyl acetate, and the combined organic layer was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The solvent was distilled off, and the resulting residue was purified by NH silica gel column chromatography (ethyl acetate: methanol = 1: 0 to 97: 3 to 9: 1). The target fraction was concentrated under reduced pressure, and the precipitate was collected by filtration with a mixed solution of diethyl ether and n-hexane and washed to obtain the title compound (90 mg, 83%). A representative powder X-ray diffraction angle of the obtained compound is shown (Compound (I) -Free Form A). (2θ ± 0.2 °): 10.4 °, 10.9 °, 11.4 °, 13.5 °, 16.1 °, 19.7 °, 20.4 °, 21.5 °, 23 .3 ° and 24.3 °.
1 H-NMR Spectrum (CDCl 3 ) δ (ppm): 1.70-1.92 (4H, m), 2.15-2.24 (2H, m), 2.53-2.65 (3H, m), 3.01-3.09 (5H, m), 3.26 (3H, s), 3.56-3.60 (2H, m), 3.64 (2H, t, J = 5.2 Hz), 4.15-4.20 (2H, m), 5.49-5.54 (1H, m), 6.55 (1H, d, J = 3.7 Hz), 6.61 (1H, dd, J = 5.8, 2.3 Hz), 7.24-7.28 (1H, m), 7.30-7.35 (3H, m), 7.81 (2H, d, J = 8.2 Hz ), 7.91 (1H, d, J = 2.4 Hz), 8.01 (1H, s), 8.10 (1H, d, J = 5.9 Hz), 8.50 (1H, brs).

[実施例1]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩(別名称:5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド=ブタンジオアート (2:3))の結晶の調製
[Example 1] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 succinate (other name: 5-({2-[({4- [1- (2-hydroxyethyl) piperidine-4- Yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide butanedioate (2: 3)) Preparation

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 2.93gをナスフラスコに量り取り、60mLのエタノールを加え、攪拌しながらオイルバスで70℃に加熱し、溶解させた。コハク酸(1.23g)を加え、オイルバスを切り、徐冷した。室温で約2時間攪拌した後、さらに5℃で1時間攪拌した。固体をろ取して、標記化合物を得た(3.70g)。
1H-NMR Spectrum (600MHz, CD3OD) δ(ppm): 1.96-2.10 (4H, m), 2.52 (6H, s), 2.93 (1H, m), 2.96 (3H, s), 3.01 (2H, m), 3.16 (2H, t, J=5.4 Hz), 3.22 (3H, s), 3.56 (2H, t, J=4.7 Hz), 3.61 (2H, m), 3.87 (2H, t, J=5.4 Hz), 4.14 (2H, t, J=4.6 Hz), 6.61 (1H, d, J=3.6 Hz), 6.68 (1H, dd, J=5.8, 2.3 Hz), 7.37 (1H, s), 7.42 (2H, d, J=8.3 Hz), 7.58 (1H, d, J=3.6 Hz), 7.73 (1H, d, J=2.2 Hz), 7.88 (2H, d, J=8.3 Hz), 8.08 (1H, s), 8.15 (1H, d, J=5.8 Hz).
13C-NMR(100MHz,solid state) δ(ppm): 27.1, 28.3, 29.7, 34.8, 38.0, 41.3, 54.0, 57.3, 59.7, 60.9, 72.1, 72.5, 103.3, 104.2, 108.5, 116.9, 126.9, 128.6, 134.5, 136.7, 140.7, 149.4, 151.3, 155.1, 169.5, 170.1, 175.6, 179.9, 183.7.
5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 -(2-Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 2.93 g was weighed into an eggplant flask, 60 mL of ethanol was added, and the mixture was heated to 70 ° C. in an oil bath with stirring and dissolved. . Succinic acid (1.23 g) was added, and the oil bath was cut and cooled slowly. After stirring at room temperature for about 2 hours, the mixture was further stirred at 5 ° C. for 1 hour. The solid was collected by filtration to give the title compound (3.70 g).
1 H-NMR Spectrum (600MHz, CD 3 OD) δ (ppm): 1.96-2.10 (4H, m), 2.52 (6H, s), 2.93 (1H, m), 2.96 (3H, s), 3.01 (2H , m), 3.16 (2H, t, J = 5.4 Hz), 3.22 (3H, s), 3.56 (2H, t, J = 4.7 Hz), 3.61 (2H, m), 3.87 (2H, t, J = 5.4 Hz), 4.14 (2H, t, J = 4.6 Hz), 6.61 (1H, d, J = 3.6 Hz), 6.68 (1H, dd, J = 5.8, 2.3 Hz), 7.37 (1H, s), 7.42 (2H, d, J = 8.3 Hz), 7.58 (1H, d, J = 3.6 Hz), 7.73 (1H, d, J = 2.2 Hz), 7.88 (2H, d, J = 8.3 Hz), 8.08 (1H , s), 8.15 (1H, d, J = 5.8 Hz).
13 C-NMR (100 MHz, solid state) δ (ppm): 27.1, 28.3, 29.7, 34.8, 38.0, 41.3, 54.0, 57.3, 59.7, 60.9, 72.1, 72.5, 103.3, 104.2, 108.5, 116.9, 126.9, 128.6 , 134.5, 136.7, 140.7, 149.4, 151.3, 155.1, 169.5, 170.1, 175.6, 179.9, 183.7.

[実施例2]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 0.5コハク酸塩の結晶(α)の調製 [Example 2] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide Preparation of 0.5 Succinate Crystals (α)

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド117mgおよびコハク酸(11.8mg)を30mLのナスフラスコに添加し、イソプロパノール/水(8/2,v/v)の溶液を2mL添加し、超音波照射後、室温で2〜3時間攪拌した。これをろ取し、標記化合物を得た(77.5mg)。
1H-NMR Spectrum (600MHz, CD3OD) δ(ppm): 1.86-2.00 (4H, m), 2.51 (2H, s), 2.62 (2H, m), 2.79 (1H, m), 2.87 (2H, t, J=5.5 Hz), 2.96 (3H, s), 3.22 (3H, s), 3.36 (2H, d, J=11.8 Hz), 3.56 (2H, t, J=4.6 Hz), 3.79 (2H, t, J=5.7 Hz), 4.15 (2H, t, J=4.6 Hz), 6.61 (1H, d, J=3.6 Hz), 6.68 (1H, dd, J=5.7, 2.1 Hz), 7.37 (1H, s), 7.40 (2H, d, J=8.2 Hz), 7.58 (1H, d, J=3.6 Hz), 7.73 (1H, d, J=2.0 Hz), 7.86 (2H, d, J=8.3 Hz), 8.08 (1H, s), 8.14 (1H, d, J=5.8 Hz).
5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 117 mg of-(2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and succinic acid (11.8 mg) were added to a 30 mL eggplant flask and isopropanol / water (8/2, v / v) 2 mL of the solution was added, and after ultrasonic irradiation, the mixture was stirred at room temperature for 2 to 3 hours. This was collected by filtration to give the title compound (77.5 mg).
1 H-NMR Spectrum (600MHz, CD 3 OD) δ (ppm): 1.86-2.00 (4H, m), 2.51 (2H, s), 2.62 (2H, m), 2.79 (1H, m), 2.87 (2H , t, J = 5.5 Hz), 2.96 (3H, s), 3.22 (3H, s), 3.36 (2H, d, J = 11.8 Hz), 3.56 (2H, t, J = 4.6 Hz), 3.79 (2H , t, J = 5.7 Hz), 4.15 (2H, t, J = 4.6 Hz), 6.61 (1H, d, J = 3.6 Hz), 6.68 (1H, dd, J = 5.7, 2.1 Hz), 7.37 (1H , s), 7.40 (2H, d, J = 8.2 Hz), 7.58 (1H, d, J = 3.6 Hz), 7.73 (1H, d, J = 2.0 Hz), 7.86 (2H, d, J = 8.3 Hz) ), 8.08 (1H, s), 8.14 (1H, d, J = 5.8 Hz).

[実施例3]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド マレイン酸塩の結晶の調製 [Example 3] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 Preparation of crystals of -methoxyethoxy) -N-methyl-1H-indole-1-carboxamide maleate

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 101mgにマレイン酸(24.1mg)および2mLのアセトンを加え、室温で一晩攪拌した。固体をろ取し、標記化合物を得た(113mg)。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 -(2-Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide Maleic acid (24.1 mg) and 2 mL of acetone were added to 101 mg, and the mixture was stirred at room temperature overnight. The solid was collected by filtration to give the title compound (113 mg).

[実施例4]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 0.5コハク酸塩の結晶(α)の調製 [Example 4] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide Preparation of 0.5 Succinate Crystals (α)

5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 550mg、コハク酸55.3mgおよび水5.5mL試験管に添加し、超音波照射後、室温にて一晩攪拌した。フィルターを用いて一晩かけてろ取した。メノウ乳鉢で粉砕後、40℃/75%RHで約1.5時間保存し、表題の化合物を得た(620mg)。
1H-NMR Spectrum (CD3OD) δ(ppm): 1.86-2.00 (4H, m), 2.51 (2H, s), 2.62 (2H, m), 2.79 (1H, m), 2.87 (2H, brt, J=5.5 Hz), 2.96 (3H, s), 3.22 (3H, s), 3.36 (2H, brd, J=11.8 Hz), 3.56 (2H, brt, J=4.6 Hz), 3.79 (2H, t, J=5.7 Hz), 4.15 (2H, brt, J=4.6 Hz), 6.61 (1H, d, J=3.6 Hz), 6.68 (1H, dd, J=5.7, 2.1 Hz), 7.37 (1H, s), 7.40 (2H, d, J=8.2 Hz), 7.58 (1H, d, J=3.6 Hz), 7.73 (1H, d, J=2.0 Hz), 7.86 (2H, d, J=8.3 Hz), 8.08 (1H, s), 8.14 (1H, d, J=5.8 Hz)
5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy)- N-methyl-1H-indole-1-carboxamide 550 mg, succinic acid 55.3 mg, and water 5.5 mL were added to a test tube, and the mixture was stirred overnight at room temperature after ultrasonic irradiation. Filtered overnight using a filter. After grinding in an agate mortar, it was stored at 40 ° C./75% RH for about 1.5 hours to obtain the title compound (620 mg).
1 H-NMR Spectrum (CD 3 OD) δ (ppm): 1.86-2.00 (4H, m), 2.51 (2H, s), 2.62 (2H, m), 2.79 (1H, m), 2.87 (2H, brt , J = 5.5 Hz), 2.96 (3H, s), 3.22 (3H, s), 3.36 (2H, brd, J = 11.8 Hz), 3.56 (2H, brt, J = 4.6 Hz), 3.79 (2H, t , J = 5.7 Hz), 4.15 (2H, brt, J = 4.6 Hz), 6.61 (1H, d, J = 3.6 Hz), 6.68 (1H, dd, J = 5.7, 2.1 Hz), 7.37 (1H, s ), 7.40 (2H, d, J = 8.2 Hz), 7.58 (1H, d, J = 3.6 Hz), 7.73 (1H, d, J = 2.0 Hz), 7.86 (2H, d, J = 8.3 Hz), 8.08 (1H, s), 8.14 (1H, d, J = 5.8 Hz)

[実施例5]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 0.5コハク酸塩の結晶(β)の調製 Example 5 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide Preparation of 0.5 succinate crystals (β)

実施例4で得られた試料を減圧下にて3日間乾燥させ、表題の化合物を得た。
1H-NMR Spectrum (CD3OD) δ(ppm): 1.87-2.00 (4H, m), 2.51 (2H, s), 2.65 (2H, m), 2.79 (1H, m), 2.89 (2H, brt, J=5.6 Hz), 2.95 (3H, s), 3.22 (3H, s), 3.38 (2H, brd, J=12.1 Hz), 3.56 (2H, m), 3.80 (2H, t, J=5.6 Hz), 4.14 (2H, m), 6.60 (1H, d, J=3.7 Hz), 6.67 (1H, dd, J=5.8, 2.3 Hz), 7.37 (1H, s), 7.40 (2H, d, J=8.4 Hz), 7.57 (1H, d, J=3.7 Hz), 7.73 (1H, d, J=2.3 Hz), 7.86 (2H, d, J=8.4 Hz), 8.08 (1H, s), 8.14 (1H, d, J=5.8 Hz)
The sample obtained in Example 4 was dried under reduced pressure for 3 days to give the title compound.
1 H-NMR Spectrum (CD 3 OD) δ (ppm): 1.87-2.00 (4H, m), 2.51 (2H, s), 2.65 (2H, m), 2.79 (1H, m), 2.89 (2H, brt , J = 5.6 Hz), 2.95 (3H, s), 3.22 (3H, s), 3.38 (2H, brd, J = 12.1 Hz), 3.56 (2H, m), 3.80 (2H, t, J = 5.6 Hz ), 4.14 (2H, m), 6.60 (1H, d, J = 3.7 Hz), 6.67 (1H, dd, J = 5.8, 2.3 Hz), 7.37 (1H, s), 7.40 (2H, d, J = 8.4 Hz), 7.57 (1H, d, J = 3.7 Hz), 7.73 (1H, d, J = 2.3 Hz), 7.86 (2H, d, J = 8.4 Hz), 8.08 (1H, s), 8.14 (1H , d, J = 5.8 Hz)

[実施例6]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩の非晶質の調製 [Example 6] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 Preparation of amorphous succinate

5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩 251mgを50%tert-ブチルアルコール水溶液25mLに溶解させた。試験管に溶液3mLを添加し、ドライアイスで冷却したエタノールで凍結させた。凍結乾燥機で溶媒を留去し、表題の化合物を得た(30.8mg)。
1H-NMR Spectrum (CD3OD) δ(ppm): 1.96-2.11 (4H, m), 2.53 (6H, s), 2.93 (1H, m), 2.96 (3H, s), 3.00 (2H, m), 3.15 (2H, t, J=5.4 Hz), 3.22 (3H, s), 3.56 (2H, m), 3.60 (2H, brd, J=12.4 Hz), 3.87 (2H, t, J=5.5 Hz), 4.15 (2H, brt, J=4.6 Hz), 6.61 (1H, d, J=3.7 Hz), 6.68 (1H, brd, J=3.8 Hz), 7.37 (1H, s), 7.42 (2H, d, J=8.3 Hz), 7.58 (1H, d, J=3.8 Hz), 7.73 (1H, brs), 7.88 (2H, d, J=8.3 Hz), 8.08 (1H, s), 8.15 (1H, brd, J=5.0 Hz)
5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy)- 251 mg of N-methyl-1H-indole-1-carboxamide 1.5 succinate was dissolved in 25 mL of 50% aqueous tert-butyl alcohol. 3 mL of the solution was added to the test tube and frozen with ethanol cooled with dry ice. The solvent was removed by lyophilization to give the title compound (30.8 mg).
1 H-NMR Spectrum (CD 3 OD) δ (ppm): 1.96-2.11 (4H, m), 2.53 (6H, s), 2.93 (1H, m), 2.96 (3H, s), 3.00 (2H, m ), 3.15 (2H, t, J = 5.4 Hz), 3.22 (3H, s), 3.56 (2H, m), 3.60 (2H, brd, J = 12.4 Hz), 3.87 (2H, t, J = 5.5 Hz) ), 4.15 (2H, brt, J = 4.6 Hz), 6.61 (1H, d, J = 3.7 Hz), 6.68 (1H, brd, J = 3.8 Hz), 7.37 (1H, s), 7.42 (2H, d , J = 8.3 Hz), 7.58 (1H, d, J = 3.8 Hz), 7.73 (1H, brs), 7.88 (2H, d, J = 8.3 Hz), 8.08 (1H, s), 8.15 (1H, brd , J = 5.0 Hz)

[参考例1]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドの結晶(Free Form B)の調製 [Reference Example 1] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide crystals (Free Form B)

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 93.2mgを試験管に秤取し、イソプロパノール2.99mLおよび水264μLを添加した。70〜100℃のオイルバスで加熱し、試料を溶解させた。この溶液を−5℃の恒温槽で約16時間攪拌した。析出した固体をろ取後、一晩減圧乾燥し、標記化合物を得た。得られた化合物の代表的な粉末X線回折角度を示す。(2θ±0.2°):7.8°、10.8°、13.1°、14.2°、17.8°、21.5°、21.7°、23.4°、24.5°、および29.0°。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 93.2 mg of-(2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide was weighed into a test tube, and 2.99 mL of isopropanol and 264 μL of water were added. The sample was dissolved by heating in an oil bath at 70 to 100 ° C. This solution was stirred for about 16 hours in a thermostatic bath at -5 ° C. The precipitated solid was collected by filtration and dried under reduced pressure overnight to obtain the title compound. The representative powder X-ray diffraction angle of the obtained compound is shown. (2θ ± 0.2 °): 7.8 °, 10.8 °, 13.1 °, 14.2 °, 17.8 °, 21.5 °, 21.7 °, 23.4 °, 24 .5 ° and 29.0 °.

[参考例2]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド水和物の結晶(Free Form Hydrate)の調製 [Reference Example 2] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide hydrate crystals (Free Form Hydrate)

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 208mgにイソプロパノール2mLおよび水2mLを添加した。氷水中で超音波照射後、約5℃で3日間攪拌した。懸濁した固体をろ取し、標記化合物を得た(106mg)。得られた化合物の代表的な粉末X線回折角度を示す。(2θ±0.2°):8.8°、9.6°、15.2°、16.3°、20.0°、20.8°、21.4°、22.0°、23.8°、および27.1°。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 To 208 mg of-(2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide was added 2 mL of isopropanol and 2 mL of water. After ultrasonic irradiation in ice water, the mixture was stirred at about 5 ° C. for 3 days. The suspended solid was collected by filtration to give the title compound (106 mg). The representative powder X-ray diffraction angle of the obtained compound is shown. (2θ ± 0.2 °): 8.8 °, 9.6 °, 15.2 °, 16.3 °, 20.0 °, 20.8 °, 21.4 °, 22.0 °, 23 .8 ° and 27.1 °.

[参考例3]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド メシル酸塩の結晶の調製 [Reference Example 3] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 Preparation of crystals of -methoxyethoxy) -N-methyl-1H-indole-1-carboxamide mesylate

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 30.1mgに2mLのアセトンを添加後、メシル酸(4.0μL)を加え、室温で4日間攪拌した。固体をろ取し、標記化合物を得た(20.4mg)。得られた化合物の代表的な粉末X線回折角度を示す。(2θ±0.2°):11.7°、13.7°、15.2°、16.9°、18.0°、18.7°、19.9°、21.1°、22.0°、および24.1°。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 -(2-Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide After adding 2 mL of acetone to 30.1 mg, mesylic acid (4.0 μL) was added, and the mixture was stirred at room temperature for 4 days. The solid was collected by filtration to give the title compound (20.4 mg). The representative powder X-ray diffraction angle of the obtained compound is shown. (2θ ± 0.2 °): 11.7 °, 13.7 °, 15.2 °, 16.9 °, 18.0 °, 18.7 °, 19.9 °, 21.1 °, 22 0.0 ° and 24.1 °.

[参考例4]5−((2−(4−(1−(2−ヒドロキシエチル)ピペリジン−4−イル)ベンズアミド)ピリジン−4−イル)オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド トシル酸塩の結晶の調製 [Reference Example 4] 5-((2- (4- (1- (2-hydroxyethyl) piperidin-4-yl) benzamido) pyridin-4-yl) oxy) -6- (2-methoxyethoxy) -N -Methyl-1H-indole-1-carboxamide tosylate crystal preparation

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 30.7mgに2mLのアセトンを添加後、p−トルエンスルホン酸一水和物(12.3mg)を加え、室温で4日間攪拌した。固体をろ取し、標記化合物を得た(15.1mg)。得られた化合物の代表的な粉末X線回折角度を示す。(2θ±0.2°):11.9°、12.6°、13.5°、13.8°、17.6°、18.0°、18.6°、20.4°、21.4°、および23.3°。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 -(2-Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide After adding 2 mL of acetone to 30.7 mg, p-toluenesulfonic acid monohydrate (12.3 mg) was added and Stir for days. The solid was collected by filtration to give the title compound (15.1 mg). The representative powder X-ray diffraction angle of the obtained compound is shown. (2θ ± 0.2 °): 11.9 °, 12.6 °, 13.5 °, 13.8 °, 17.6 °, 18.0 °, 18.6 °, 20.4 °, 21 .4 ° and 23.3 °.

[参考例5]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 安息香酸塩の結晶の調製 [Reference Example 5] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 -Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide Preparation of benzoate crystals

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 20.3mgおよび安息香酸(8.91mg)の混合物に0.2mLの酢酸エチルを加え、室温にて攪拌した。2時間後、0.1mLの酢酸エチルを追加し、さらに一晩攪拌した。固体をろ取し、標記化合物を得た。得られた化合物の代表的な粉末X線回折角度を示す。(2θ±0.2°):9.3°、13.9°、14.5°、15.8°、18.1°、19.4°、20.5°、21.3°、22.6°、および26.2°。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 0.2 mL of ethyl acetate was added to a mixture of-(2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 20.3 mg and benzoic acid (8.91 mg), and the mixture was stirred at room temperature. After 2 hours, 0.1 mL of ethyl acetate was added, and the mixture was further stirred overnight. The solid was collected by filtration to give the title compound. The representative powder X-ray diffraction angle of the obtained compound is shown. (2θ ± 0.2 °): 9.3 °, 13.9 °, 14.5 °, 15.8 °, 18.1 °, 19.4 °, 20.5 °, 21.3 °, 22 .6 ° and 26.2 °.

[参考例6]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド フマル酸塩の結晶の調製 [Reference Example 6] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 Preparation of crystals of -methoxyethoxy) -N-methyl-1H-indole-1-carboxamide fumarate

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 30.6mgおよびフマル酸(7.24mg)の混合物に2mLのアセトンを添加し、室温にて一晩攪拌した。固体をろ取し、標記化合物を得た。得られた化合物の代表的な粉末X線回折角度を示す。(2θ±0.2°):9.6°、13.8°、15.7°、16.7°、19.8°、21.0°、22.0°、22.4°、24.7°、および25.7°。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 2 mL of acetone was added to a mixture of 30.6 mg of-(2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and fumaric acid (7.24 mg) and stirred overnight at room temperature. The solid was collected by filtration to give the title compound. The representative powder X-ray diffraction angle of the obtained compound is shown. (2θ ± 0.2 °): 9.6 °, 13.8 °, 15.7 °, 16.7 °, 19.8 °, 21.0 °, 22.0 °, 22.4 °, 24 .7 ° and 25.7 °.

[参考例7]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 塩酸塩の調製 [Reference Example 7] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 Preparation of -methoxyethoxy) -N-methyl-1H-indole-1-carboxamide hydrochloride

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 29.5mgに2mLのアセトンおよび6N塩酸(10.0μL)を添加し、室温にて攪拌したところ、標記化合物が、溶媒から油状物質として分離した。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 -(2-Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 29.5 mg of acetone and 6N hydrochloric acid (10.0 μL) were added to 29.5 mg and the mixture was stirred at room temperature. As an oil.

[参考例8]5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 臭化水素酸塩の調製 [Reference Example 8] 5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2 Preparation of -methoxyethoxy) -N-methyl-1H-indole-1-carboxamide hydrobromide

製造例1−15に記載の5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 32.7mgに2mLのアセトンおよび臭化水素酸(7.8μL)を添加し、室温にて攪拌したところ、標記化合物が、溶媒から油状物質として分離した。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6 described in Preparation Example 1-15 2 mL of acetone and hydrobromic acid (7.8 μL) were added to 32.7 mg of-(2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide, and the mixture was stirred at room temperature. Separated from the solvent as an oil.

[試験例]
以下の試験例を行い、製造例1−15記載の化合物(I)もしくは化合物(I)の塩またはその結晶の、物性または薬理効果を調べた。
[Test example]
The following test examples were conducted to examine the physical properties or pharmacological effects of the compound (I) or the salt of compound (I) described in Production Example 1-15 or crystals thereof.

[試験例1]吸湿性
動的水分吸着測定装置により、実施例1の化合物(I)・1.5コハク酸塩の吸湿性を評価した。装置の試料装着部位を25℃に保温し、相対湿度(RH)を5%〜95%の範囲で段階的に設定した。湿度は0%RHの乾燥窒素および100%RHの加湿窒素の相対流量を変えることで調節した。試料重量は2分間隔ごとにミクロ天秤で確認され、5分間の重量変動幅が0.01%を下回った時点で順次湿度を変更した。その結果を図5に示す。
[Test Example 1] Hygroscopicity The hygroscopicity of the compound (I) · 1.5 succinate of Example 1 was evaluated using a dynamic moisture adsorption measuring apparatus. The sample mounting part of the apparatus was kept at 25 ° C., and the relative humidity (RH) was set stepwise in the range of 5% to 95%. Humidity was adjusted by changing the relative flow rates of 0% RH dry nitrogen and 100% RH humidified nitrogen. The sample weight was confirmed with a microbalance every 2 minutes, and the humidity was sequentially changed when the weight fluctuation range for 5 minutes was less than 0.01%. The result is shown in FIG.

[試験例2]セルフリーキナーゼ阻害作用
平底96ウェル白プレート(住友ベークライト MS−8496W)に、アッセイバッファー(20mM HEPES−NaOH、0.01% TritonX−100、2mM DTT、5mM MgCl)にて1μg/mLに希釈したFGFR1蛋白(カルナバイオサイエンス社 08−133)溶液10μl、最終濃度1000nMのCSK−tide substrate(AnaSpec Inc 63843)および最終濃度58.3μMのATP(promega V9102)を含むアッセイバッファー溶液10μL、アッセイバッファーにて希釈した被験物質5μlを加え、室温で1時間反応させた。キナーゼ活性測定にはADP−GloTM Kinase Assay(promega V9102)を用いた。反応後のプレートにADP−Glo reagentを各ウェル25μL添加し、室温で40分間反応させキナーゼ反応を停止し、残存ATPを枯渇させた。さらにKinase detection reagentを加え、室温で40分間反応させADPからATPへの変換とルシフェラーゼ/ルシフェリンカップリング反応とATPによる発光反応を行った。EnvisionTM(パーキンエルマー株式会社)にて各ウェルの発光量を測定し酵素活性を評価した。被験物質を添加せずにキナーゼタンパク質を添加した場合の発光量を100%、被験物質およびキナーゼタンパク質を添加していない場合の発光量を0%として、被験物質存在下での発光量率を求めた。この発光量率によりキナーゼ活性を50%阻害するのに必要な被験物質の濃度(IC50値)を算出した。
[Test Example 2] Cell-free kinase inhibitory action 1 μg of assay solution (20 mM HEPES-NaOH, 0.01% Triton X-100, 2 mM DTT, 5 mM MgCl 2 ) on a flat bottom 96 well white plate (Sumitomo Bakelite MS-8396W) 10 μL of an assay buffer solution containing 10 μl of FGFR1 protein (Carna Biosciences 08-133) diluted to / mL, final concentration of 1000 nM CSK-tide substrate (AnaSpec Inc 633843) and final concentration of 58.3 μM ATP (promega V9102) Then, 5 μl of a test substance diluted with an assay buffer was added and reacted at room temperature for 1 hour. ADP-Glo Kinase Assay (promega V9102) was used for the kinase activity measurement. To the plate after the reaction, 25 μL of ADP-Glo reagent was added to each well and reacted at room temperature for 40 minutes to stop the kinase reaction and deplete the remaining ATP. Further, Kinase detection reagent was added and reacted at room temperature for 40 minutes to perform conversion from ADP to ATP, luciferase / luciferin coupling reaction, and luminescence reaction by ATP. The enzyme activity was evaluated by measuring the amount of luminescence in each well using Envision (Perkin Elmer Co., Ltd.). Calculate the luminescence rate in the presence of the test substance, assuming that the luminescence when the kinase protein is added without adding the test substance is 100%, and the luminescence when the test substance and kinase protein are not added is 0%. It was. The concentration (IC 50 value) of the test substance necessary to inhibit the kinase activity by 50% was calculated from this luminescence amount rate.

前記FGFR1セルフリーキナーゼ阻害作用と同様にして、FGFR2蛋白(カルナバイオサイエンス社 08−134)、FGFR3蛋白(カルナバイオサイエンス社 08−135)、またはFGFR4蛋白(カルナバイオサイエンス社 08−136)を用いて、FGFR2セルフリーキナーゼ阻害作用、FGFR3セルフリーキナーゼ阻害作用、およびFGFR4セルフリーキナーゼ阻害作用をそれぞれ測定した。ただし、ATP濃度に関してはFGFR2では最終濃度35μM、FGFR3では最終濃度16.7μM、FGFR4では最終濃度75μMにて評価した。またFGFR3およびFGFR4に関して、被験物質との反応時間は室温2時間とした。これらの結果を表1に示した。   Similar to the FGFR1 cell-free kinase inhibitory action, FGFR2 protein (Karna Biosciences 08-134), FGFR3 protein (Karna Biosciences 08-135), or FGFR4 protein (Karna Biosciences 08-136) was used. The FGFR2 cell-free kinase inhibitory action, the FGFR3 cell-free kinase inhibitory action, and the FGFR4 cell-free kinase inhibitory action were each measured. However, the ATP concentration was evaluated at a final concentration of 35 μM for FGFR2, 16.7 μM for FGFR3, and 75 μM for FGFR4. Regarding FGFR3 and FGFR4, the reaction time with the test substance was 2 hours at room temperature. These results are shown in Table 1.

<セルフリーキナーゼ阻害作用データ>
<Cell-free kinase inhibitory action data>

[試験例3]SNU−16増殖阻害アッセイ
ヒト胃がん細胞株SNU−16(ATCC Number CRL−5974)はFGFR2遺伝子増幅を有することが報告されている(Cancer Res.2008.68:2340−2348)。SNU−16細胞は10% FBS、ペニシリン/ストレプトマイシン(WAKO 168−23191)を含むRPMI−1640(WAKO 187−02021)培地を用い、5%COインキュベーター中(37℃)で培養維持を行った。96ウェルプレート(ベクトンディッキンソン 35−3075)の各ウェルに、10%FBSを含むRPMI−1640培地を用いて1×10 cells/mLに調製したSNU−16の細胞懸濁液を150μLずつ加え、5%COインキュベーター中(37℃)で一晩培養した。翌日、10%FBSを含むRPMI−1640培地にて希釈した被験物質を50μLずつ添加し、5%COインキュベーター中(37℃)で3日間培養した。Cell Counting Kit−8(同仁化学研究所 CK04)10μLを各ウェルに添加し、5%COインキュベーター中(37℃)で1−2時間培養し発色させた。ENVISIONTM(パーキンエルマー社)により450nmの吸光度を測定した。被験物質を添加していない場合の吸光度を100%、細胞が存在していないウェルの吸光度を0%として、被験物質存在下での吸光度率を求めた。細胞増殖を50%阻害するのに必要な被験物質の濃度(IC50値)を求め、表2に示した。
[Test Example 3] SNU-16 Growth Inhibition Assay It has been reported that human gastric cancer cell line SNU-16 (ATCC Number CRL-5974) has FGFR2 gene amplification (Cancer Res. 2008.68: 2340-2348). SNU-16 cells were cultured and maintained in a 5% CO 2 incubator (37 ° C.) using RPMI-1640 (WAKO 187-02021) medium containing 10% FBS and penicillin / streptomycin (WAKO 168-23191). To each well of a 96-well plate (Becton Dickinson 35-3075), 150 μL each of SNU-16 cell suspension prepared to 1 × 10 4 cells / mL using RPMI-1640 medium containing 10% FBS was added. The cells were cultured overnight in a 5% CO 2 incubator (37 ° C.). On the next day, 50 μL each of the test substance diluted in RPMI-1640 medium containing 10% FBS was added and cultured in a 5% CO 2 incubator (37 ° C.) for 3 days. 10 μL of Cell Counting Kit-8 (Dojindo Laboratories CK04) was added to each well, and the cells were cultured in a 5% CO 2 incubator (37 ° C.) for 1-2 hours for color development. Absorbance at 450 nm was measured by ENVISION (Perkin Elmer). Absorbance rate in the presence of the test substance was determined with the absorbance in the absence of the test substance being 100% and the absorbance of the well in which no cells were present being 0%. The concentration (IC 50 value) of the test substance necessary to inhibit cell growth by 50% was determined and shown in Table 2.

<SNU−16増殖抑制作用評価データ>
<SNU-16 growth inhibitory action evaluation data>

[試験例4]マウスSNU−16皮下移植モデルにおける抗腫瘍効果
10%FBS、ペニシリン/ストレプトマイシンを含むRPMI−1640培養液で培養したヒト胃がん細胞株SNU−16をHanks’ Balanced Salt Solution (GIBCO #24020)にて1×10 cells/mL濃度に調製し、MATRIGEL(BDバイオサイエンス Cat# 354234)と1:1で混合し5×10 cells/mlの細胞懸濁液を調製した。6〜7週齢のヌードマウス(BALB/cAJcl−nu/nu、雌、日本クレア株式会社)の右脇腹皮下部に100μLの容量で移植した。移植から7日後に電子デジタルノギス(デジマチックTMキャリパ、株式会社ミツトヨ)を用いて腫瘍の短径、長径を計測し、以下の計算式で腫瘍体積を算出した。
腫瘍体積(mm)=長径(mm)×短径(mm)×短径(mm)/2
投与初日の腫瘍体積をもとに腫瘍体積の平均値がほぼ等しくなるように群分けを行った。被験物質はDMSOに溶解し、Tween80を加え、10倍濃度の溶液を調製し冷凍保存した。投与直前に5%グルコース溶液を添加し、最終投与溶液とした(DMSO:Tween80:5% glucose溶液=3.5%:6.5%:90%)。評価検体は20mL/kgの投与容量で1日1回11日間連続経口投与し、コントロール群には投与溶媒を同条件で経口投与した。尚、実験は1群5匹で行った。
コントロール群、被験物質投与群それぞれに対し、初日の体重に対する最終日の体重比(relative body weight: RBW)を算出する。被験物質投与群のRBW/コントロール群のRBWが0.9の以上の被験物質投与群を安全に投与可能な群と判定した。これに該当する被験物質投与群について、最終日におけるコントロールの腫瘍体積に対する被験物質投与後の腫瘍体積の割合(T/C)(%)を算出し、表3に示した。
Test Example 4 Antitumor Effect in Mouse SNU-16 Subcutaneous Transplant Model Human gastric cancer cell line SNU-16 cultured in RPMI-1640 culture solution containing 10% FBS and penicillin / streptomycin was treated with Hanks' Balanced Salt Solution (GIBCO # 24020 ) To a concentration of 1 × 10 8 cells / mL and mixed 1: 1 with MATRIGEL (BD Bioscience Cat # 354234) to prepare a cell suspension of 5 × 10 7 cells / ml. A nude mouse (BALB / cAJcl-nu / nu, female, Nippon Claire Co., Ltd.) of 6-7 weeks old was transplanted into the right flank subcutaneous part in a volume of 100 μL. Seven days after transplantation, the short axis and long axis of the tumor were measured using an electronic digital caliper (Digimatic TM caliper, Mitutoyo Corporation), and the tumor volume was calculated by the following formula.
Tumor volume (mm 3 ) = major axis (mm) × minor axis (mm) × minor axis (mm) / 2
Based on the tumor volume on the first day of administration, the groups were divided so that the average values of the tumor volumes were almost equal. The test substance was dissolved in DMSO, Tween 80 was added, a 10-fold concentrated solution was prepared, and stored frozen. Immediately before administration, a 5% glucose solution was added to obtain a final administration solution (DMSO: Tween 80: 5% glucose solution = 3.5%: 6.5%: 90%). The test sample was orally administered once a day for 11 days at a dose volume of 20 mL / kg, and the control solvent was orally administered under the same conditions. The experiment was conducted with 5 animals per group.
For each of the control group and the test substance-administered group, the weight ratio (RBW) of the last day to the weight of the first day is calculated. The test substance administration group in which the RBW of the test substance administration group / RBW of the control group was 0.9 or more was determined to be a safe administration group. For the test substance administration group corresponding to this, the ratio (T / C) (%) of the tumor volume after test substance administration to the control tumor volume on the last day was calculated and shown in Table 3.

<SNU−16皮下移植モデルにおける抗腫瘍効果評価データ>
<Anti-tumor effect evaluation data in SNU-16 subcutaneous transplantation model>

Claims (15)

下記式(I)で表される5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドと、コハク酸またはマレイン酸からなる塩。
5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy)-represented by the following formula (I) A salt comprising 6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and succinic acid or maleic acid.
コハク酸塩である、請求項1記載の塩。   The salt of claim 1 which is a succinate. 5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩である、請求項2記載の塩。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy)- The salt of claim 2 which is N-methyl-1H-indole-1-carboxamide 1.5 succinate. 5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 0.5コハク酸塩である、請求項2記載の塩。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy)- The salt of claim 2 which is N-methyl-1H-indole-1-carboxamide 0.5 succinate. 5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド マレイン酸塩である、請求項1記載の塩。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy)- The salt according to claim 1, which is N-methyl-1H-indole-1-carboxamide maleate. 下記式(I)で表される5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドと、コハク酸からなる塩の結晶。
5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy)-represented by the following formula (I) Crystal of salt consisting of 6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and succinic acid.
下記式(I)で表される5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミドと、マレイン酸からなる塩の結晶。
5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy)-represented by the following formula (I) Crystal of salt consisting of 6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide and maleic acid.
粉末X線回折において、回折角度(2θ±0.2°)22.4°に回折ピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩の結晶。   In powder X-ray diffraction, 5-({2-[({4- [1- (2-hydroxyethyl) piperidine-4-] has a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 22.4 °. Yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 succinate crystals. 粉末X線回折において、回折角度(2θ±0.2°)22.4°、25.3°および23.3°に回折ピークを有する、請求項8記載の結晶。   The crystal according to claim 8, having diffraction peaks at diffraction angles (2θ ± 0.2 °) of 22.4 °, 25.3 °, and 23.3 ° in powder X-ray diffraction. 粉末X線回折において、回折角度(2θ±0.2°)19.8°に回折ピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 0.5コハク酸塩の結晶(α)。   In powder X-ray diffraction, 5-({2-[({4- [1- (2-hydroxyethyl) piperidine-4-] has a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 19.8 °. Yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 0.5 succinate crystals (α). 粉末X線回折において、回折角度(2θ±0.2°)20.1°に回折ピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド マレイン酸塩の結晶。   In powder X-ray diffraction, 5-({2-[({4- [1- (2-hydroxyethyl) piperidine-4-] has a diffraction peak at a diffraction angle (2θ ± 0.2 °) of 20.1 °. Yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide maleate crystals. 13C固体NMRスペクトルにおいて、化学シフト(±0.5ppm)108.5ppm、155.1ppmおよび179.9ppmにピークを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩の結晶。 In the 13 C solid state NMR spectrum, 5-({2-[({4- [1- (2-hydroxyethyl) has peaks at chemical shifts (± 0.5 ppm) 108.5 ppm, 155.1 ppm and 179.9 ppm. ) Piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy) -6- (2-methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 Succinate crystals . 13C固体NMRスペクトルにおいて、化学シフト(±0.5ppm)27.1ppm、34.8ppm、108.5ppm、155.1ppmおよび179.9ppmにピークを有する、請求項12記載の結晶。The crystal according to claim 12, having peaks at chemical shifts (± 0.5 ppm) of 27.1 ppm, 34.8 ppm, 108.5 ppm, 155.1 ppm and 179.9 ppm in a 13 C solid state NMR spectrum. 図1の粉末X線回折パターンを有する、5−({2−[({4−[1−(2−ヒドロキシエチル)ピペリジン−4−イル]フェニル}カルボニル)アミノ]ピリジン−4−イル}オキシ)−6−(2−メトキシエトキシ)−N−メチル−1H−インドール−1−カルボキサミド 1.5コハク酸塩の結晶。   5-({2-[({4- [1- (2-hydroxyethyl) piperidin-4-yl] phenyl} carbonyl) amino] pyridin-4-yl} oxy having the powder X-ray diffraction pattern of FIG. ) -6- (2-Methoxyethoxy) -N-methyl-1H-indole-1-carboxamide 1.5 Succinate crystals. 請求項1〜14のいずれか一項に記載の塩または結晶を有効成分として含有する医薬組成物。   The pharmaceutical composition which contains the salt or crystal | crystallization as described in any one of Claims 1-14 as an active ingredient.
JP2015560425A 2014-08-18 2015-08-17 Salts and crystals of monocyclic pyridine derivatives Active JP5925978B1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2014166118 2014-08-18
JP2014166118 2014-08-18
PCT/JP2015/073047 WO2016027781A1 (en) 2014-08-18 2015-08-17 Salt of monocyclic pyridine derivative and crystal thereof

Publications (2)

Publication Number Publication Date
JP5925978B1 JP5925978B1 (en) 2016-05-25
JPWO2016027781A1 true JPWO2016027781A1 (en) 2017-04-27

Family

ID=55350725

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2015560425A Active JP5925978B1 (en) 2014-08-18 2015-08-17 Salts and crystals of monocyclic pyridine derivatives

Country Status (14)

Country Link
US (1) US9951047B2 (en)
EP (1) EP3184520B1 (en)
JP (1) JP5925978B1 (en)
KR (1) KR102344105B1 (en)
CN (1) CN106660997B (en)
AU (1) AU2015304465B2 (en)
BR (1) BR112017002268B1 (en)
CA (1) CA2956270C (en)
ES (1) ES2914072T3 (en)
IL (1) IL250290B (en)
MX (1) MX369646B (en)
RU (1) RU2658821C1 (en)
SG (1) SG11201700703XA (en)
WO (1) WO2016027781A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6858132B2 (en) * 2015-12-17 2021-04-14 エーザイ・アール・アンド・ディー・マネジメント株式会社 Breast cancer treatment
CN111050768A (en) * 2017-10-12 2020-04-21 卫材R&D管理有限公司 Pharmaceutical compositions comprising FGFR selective tyrosine kinase inhibitors
US11219619B2 (en) 2018-03-28 2022-01-11 Eisai R&D Management Co., Ltd. Therapeutic agent for hepatocellular carcinoma
MX2022012934A (en) 2020-04-17 2022-11-08 Eisai R&D Man Co Ltd Breast cancer therapeutic agent.
JPWO2022025150A1 (en) 2020-07-31 2022-02-03
EP4209216A1 (en) 2020-10-28 2023-07-12 Eisai R&D Management Co., Ltd Pharmaceutical composition for treating tumors
CN117677613A (en) 2021-08-31 2024-03-08 卫材R&D管理有限公司 Process for producing synthetic intermediate of monocyclic pyridine derivative
CN117715902A (en) 2021-08-31 2024-03-15 卫材R&D管理有限公司 Process for producing monocyclic pyridine derivative
WO2023174400A1 (en) * 2022-03-18 2023-09-21 上海润石医药科技有限公司 Salt of substituted amino six-membered nitric heterocyclic compound, crystal form thereof, method for preparing same, and use thereof

Family Cites Families (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10006139A1 (en) 2000-02-11 2001-08-16 Merck Patent Gmbh Indol-3-yl derivatives
JP3712393B2 (en) 2000-10-20 2005-11-02 エーザイ株式会社 Nitrogen-containing aromatic ring derivatives
US20030187026A1 (en) 2001-12-13 2003-10-02 Qun Li Kinase inhibitors
KR100732440B1 (en) 2002-08-30 2007-06-27 에자이 알앤드디 매니지먼트 가부시키가이샤 Azaarene derivatives
US7098332B2 (en) 2002-12-20 2006-08-29 Hoffmann-La Roche Inc. 5,8-Dihydro-6H-pyrido[2,3-d]pyrimidin-7-ones
RU2350617C2 (en) 2003-04-10 2009-03-27 Ф.Хоффманн-Ля Рош Аг Pyrimidine compounds with selective inhibition of "¦-¦" and "l¦l¦" activity
US20050256154A1 (en) 2004-05-04 2005-11-17 Kin-Chun Luk 4-Amino-thieno[3,2-c]pyridine-7-carboxylic acid amides
GB0512324D0 (en) 2005-06-16 2005-07-27 Novartis Ag Organic compounds
FR2883286B1 (en) 2005-03-16 2008-10-03 Sanofi Aventis Sa NOVEL IMIDAZO [1,5-a] PYRIDINE DERIVATIVES, INHIBITORS OF FGFs, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING SAME
EP2270000B1 (en) 2005-05-23 2015-07-29 Novartis AG Crystalline and other forms of 4-amino-5-fluoro-3-[6-(4-methylpiperazin-1-yl)-1H-benzimidazol-2-yl]-1H-quinolin-2-one lactic acid salts
SMP200800041B (en) 2005-12-21 2009-07-14 Novartis Ag Active pyrimidinyl-aryl-urea derivatives such as fgf-inhibitors
TW200811134A (en) 2006-07-12 2008-03-01 Irm Llc Compounds and compositions as protein kinase inhibitors
EP1891955A1 (en) 2006-07-24 2008-02-27 Sanofi-Aventis Use of 1,2,3-substituted indolizine derivatives, inhibitors of FGFs, for the preparation of a medicament intended for the treatment of degenerative joint diseases
US7737149B2 (en) 2006-12-21 2010-06-15 Astrazeneca Ab N-[5-[2-(3,5-dimethoxyphenyl)ethyl]-2H-pyrazol-3-yl]-4-(3,5-dimethylpiperazin-1-yl)benzamide and salts thereof
US8131527B1 (en) 2006-12-22 2012-03-06 Astex Therapeutics Ltd. FGFR pharmacophore compounds
MX2009006706A (en) 2006-12-22 2009-07-02 Astex Therapeutics Ltd Bicyclic heterocyclic compounds as fgfr inhibitors.
JP5442449B2 (en) 2006-12-22 2014-03-12 アステックス、セラピューティックス、リミテッド New compounds
EP2158470A1 (en) 2007-06-25 2010-03-03 Qinetiq Limited Preconcentrator device incorporating a polymer of intrinsic microporosity
WO2009019518A1 (en) 2007-08-09 2009-02-12 Astrazeneca Ab Pyrimidine compounds having a fgfr inhibitory effect
GB0720038D0 (en) 2007-10-12 2007-11-21 Astex Therapeutics Ltd New compounds
GB0720041D0 (en) 2007-10-12 2007-11-21 Astex Therapeutics Ltd New Compounds
WO2009056886A1 (en) 2007-11-01 2009-05-07 Astrazeneca Ab Pyrimidine derivatives and their use as modulators of fgfr activity
CL2008003675A1 (en) 2007-12-13 2009-03-20 Wyeth Corp Compounds derived from 5-alkyl or alkenyl 3-cyanopyridines, preparation process, pharmaceutical composition, useful to reduce the increased activity of an enzyme in a mammal, wherein said enzyme is a protein kinase, intended for the treatment of inflammation, asthma, colitis, multiple sclerosis, psoriasis, rheumatoid arthritis.
EA021421B1 (en) 2008-05-23 2015-06-30 Новартис Аг Derivatives of quinolines and quinoxalines as protein tyrosine kinase inhibitors, process for preparation thereof, pharmaceutical composition comprising same and method for treating diseases using such compounds
GB0810902D0 (en) 2008-06-13 2008-07-23 Astex Therapeutics Ltd New compounds
EA201100030A1 (en) 2008-06-19 2011-08-30 Астразенека Аб PYRAZOL COMPOUNDS 436
TW201035098A (en) 2008-12-30 2010-10-01 Arqule Inc Substituted imidazolyl-5,6-dihydrobenzo[n] isoquinoline compounds
AU2009335019A1 (en) 2008-12-30 2011-08-18 Arqule, Inc. Substituted 1H-pyrazolo[3,4-d]pyrimidine-6-amine compounds
EP2379506B1 (en) 2008-12-30 2015-09-02 ArQule, Inc. Substituted 5,6-dihydro-6-phenylbenzo[f]isoquinolin-2-amine compounds
US9002427B2 (en) 2009-03-30 2015-04-07 Lifewave Biomedical, Inc. Apparatus and method for continuous noninvasive measurement of respiratory function and events
GB0906470D0 (en) 2009-04-15 2009-05-20 Astex Therapeutics Ltd New compounds
GB0906472D0 (en) 2009-04-15 2009-05-20 Astex Therapeutics Ltd New compounds
EP2270043A1 (en) 2009-07-03 2011-01-05 Sanofi-Aventis Extracellular allosteric inhibitor binding domain from a tyrosine kinase receptor
FR2947546B1 (en) 2009-07-03 2011-07-01 Sanofi Aventis PYRAZOLE DERIVATIVES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
KR101529767B1 (en) 2009-08-07 2015-06-17 추가이 세이야쿠 가부시키가이샤 Aminopyrazole derivative
CN102596916B (en) 2009-10-30 2015-06-17 诺瓦提斯公司 N-oxide of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea
AR079257A1 (en) 2009-12-07 2012-01-04 Novartis Ag CRYSTAL FORMS OF 3- (2,6-DICLORO-3-5-DIMETOXI-PHENYL) -1- {6- [4- (4-ETIL-PIPERAZIN-1-IL) -PENYL-AMINO] -PIRIMIDIN-4- IL} -1-METHYL-UREA AND SALTS OF THE SAME
GB201007286D0 (en) 2010-04-30 2010-06-16 Astex Therapeutics Ltd New compounds
FR2962437B1 (en) 2010-07-06 2012-08-17 Sanofi Aventis IMIDAZOPYRIDINE DERIVATIVES, PROCESS FOR PREPARING THEM AND THEIR THERAPEUTIC APPLICATION
GB201020179D0 (en) 2010-11-29 2011-01-12 Astex Therapeutics Ltd New compounds
WO2012088266A2 (en) 2010-12-22 2012-06-28 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of fgfr3
EP2548877A1 (en) 2011-07-19 2013-01-23 MSD Oss B.V. 4-(5-Membered fused pyridinyl)benzamides as BTK-inhibitors
DK2734522T3 (en) 2011-07-19 2019-02-18 Merck Sharp & Dohme 4-IMIDAZOPYRIDAZIN-1-YL-BENZAMIDES AND 4-IMIDAZOTRIAZIN-1-YL-BENZAMIDES AS BTK INHIBITORS
GB201118675D0 (en) 2011-10-28 2011-12-14 Astex Therapeutics Ltd New compounds
GB201118654D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
GB201118652D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
GB201118656D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
FR2984325A1 (en) 2011-12-14 2013-06-21 Sanofi Sa PYRAZOLOPYRIDINE DERIVATIVES, PROCESS FOR PREPARING THEM AND THEIR THERAPEUTIC APPLICATION
MX351513B (en) 2012-01-19 2017-10-17 Taiho Pharmaceutical Co Ltd 3,5-disubstituted alkynylbenzene compound and salt thereof.
JP5343177B1 (en) 2012-02-28 2013-11-13 アステラス製薬株式会社 Nitrogen-containing aromatic heterocyclic compounds
GB201209609D0 (en) 2012-05-30 2012-07-11 Astex Therapeutics Ltd New compounds
LT3495367T (en) 2012-06-13 2021-02-25 Incyte Holdings Corporation Substituted tricyclic compounds as fgfr inhibitors
CN104619840A (en) 2012-07-05 2015-05-13 日本国立癌症研究中心 Fgfr2 fusion gene
RS62233B1 (en) 2012-07-11 2021-09-30 Blueprint Medicines Corp Inhibitors of the fibroblast growth factor receptor
WO2014026125A1 (en) 2012-08-10 2014-02-13 Incyte Corporation Pyrazine derivatives as fgfr inhibitors
WO2014044846A1 (en) 2012-09-24 2014-03-27 Evotec (Uk) Ltd. 3-(aryl- or heteroaryl-amino)-7-(3,5-dimethoxyphenyl)isoquinoline derivatives as fgfr inhibitors useful for the treatment of proliferative disorders or dysplasia
WO2014048878A1 (en) 2012-09-26 2014-04-03 Evotec (Uk) Ltd. Phenyl- or pyridyl- pyrrolo[2,3b]pyrazine derivatives useful in the treatment or prevention of proliferative disorders or dysplasia
CN104797710A (en) 2012-09-27 2015-07-22 中外制药株式会社 FGFR3 fusion gene and pharmaceutical drug targeting same
AR094812A1 (en) 2013-02-20 2015-08-26 Eisai R&D Man Co Ltd DERIVED FROM MONOCYCLIC PYRIDINE AS AN FGFR INHIBITOR
TWI628176B (en) 2013-04-04 2018-07-01 奧利安公司 Protein kinase inhibitors

Also Published As

Publication number Publication date
IL250290B (en) 2020-06-30
EP3184520B1 (en) 2022-03-23
US20170217935A1 (en) 2017-08-03
CA2956270A1 (en) 2016-02-25
BR112017002268B1 (en) 2022-11-08
IL250290A0 (en) 2017-03-30
JP5925978B1 (en) 2016-05-25
AU2015304465B2 (en) 2019-05-09
SG11201700703XA (en) 2017-03-30
MX2017001624A (en) 2017-04-27
CA2956270C (en) 2022-08-09
BR112017002268A2 (en) 2017-11-21
EP3184520A4 (en) 2018-04-18
CN106660997A (en) 2017-05-10
CN106660997B (en) 2019-05-21
KR102344105B1 (en) 2021-12-29
KR20170035927A (en) 2017-03-31
EP3184520A1 (en) 2017-06-28
RU2658821C1 (en) 2018-06-25
ES2914072T3 (en) 2022-06-07
AU2015304465A1 (en) 2017-02-23
MX369646B (en) 2019-11-15
US9951047B2 (en) 2018-04-24
WO2016027781A1 (en) 2016-02-25

Similar Documents

Publication Publication Date Title
JP5925978B1 (en) Salts and crystals of monocyclic pyridine derivatives
DK2805940T3 (en) PYRAZINE CARBOXAMIDE COMPOUND
JP5600229B1 (en) Monocyclic pyridine derivative
JP7470058B2 (en) Heterocyclic compounds
WO2015016206A1 (en) Heterocyclic compound
WO2015190564A1 (en) Nitrogen-containing heterocyclic compound
KR20200067856A (en) Epithelial growth factor receptor inhibitor
KR20170043546A (en) Quinazoline Derivative
JP2019518776A (en) Crystals of Aniline Pyrimidine Compounds as EGFR Inhibitors
WO2023060362A1 (en) Ras inhibitors, compositions and methods of use thereof
JP2021533143A (en) CDK8 / 19 inhibitor
CN114929675A (en) Novel adamantane derivatives as inhibitors of focal adhesion kinase
JP2017206437A (en) 4-aminopyridine derivative
JP2006104195A (en) Pyrimidine derivative and medicine
JP2023541140A (en) Salts of compounds for degrading BTK, their crystalline forms and their use in medicine

Legal Events

Date Code Title Description
A975 Report on accelerated examination

Free format text: JAPANESE INTERMEDIATE CODE: A971005

Effective date: 20160302

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A132

Effective date: 20160308

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20160324

TRDD Decision of grant or rejection written
A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

Effective date: 20160419

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20160420

R150 Certificate of patent or registration of utility model

Ref document number: 5925978

Country of ref document: JP

Free format text: JAPANESE INTERMEDIATE CODE: R150

R250 Receipt of annual fees

Free format text: JAPANESE INTERMEDIATE CODE: R250